WO2007124136A1 - Use of 4-amino-piperidines for treating sleep disorders - Google Patents

Use of 4-amino-piperidines for treating sleep disorders Download PDF

Info

Publication number
WO2007124136A1
WO2007124136A1 PCT/US2007/009804 US2007009804W WO2007124136A1 WO 2007124136 A1 WO2007124136 A1 WO 2007124136A1 US 2007009804 W US2007009804 W US 2007009804W WO 2007124136 A1 WO2007124136 A1 WO 2007124136A1
Authority
WO
WIPO (PCT)
Prior art keywords
methylpiperidin
carbamide
methylpropyloxy
phenylmethyl
fluorophenylmethyl
Prior art date
Application number
PCT/US2007/009804
Other languages
French (fr)
Inventor
Daniel Van Kammen
David M. Weiner
Mark R. Brann
Robert E. Davis
Original Assignee
Acadia Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Acadia Pharmaceuticals, Inc. filed Critical Acadia Pharmaceuticals, Inc.
Publication of WO2007124136A1 publication Critical patent/WO2007124136A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4468Non condensed piperidines, e.g. piperocaine having a nitrogen directly attached in position 4, e.g. clebopride, fentanyl
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system

Definitions

  • the present invention relates to treating sleep disorders using compounds that are selective towards monoamine receptors such as serotonin receptors.
  • Compounds that are useful for treating sleep disorders include 4-amino-piperidines.
  • insomnia sleep maintenance insomnia
  • SMI Sleep maintenance insomnia
  • One embodiment described herein includes a method of increasing slow- wave sleep comprising administering N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide to a subject in need of increased slow wave sleep in an amount sufficient to increase slow wave sleep.
  • One embodiment further comprises informing the subject that N-(I -methylpiperidin-4-yl)- N-(4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide increases slow wave sleep.
  • the informing comprises providing printed matter that advises that N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide increases slow wave sleep.
  • the printed matter is a label.
  • Another embodiment disclosed herein includes a method of treating insomnia comprising administering N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide to a subject suffering from insomnia in an amount sufficient to ameliorate insomnia.
  • One embodiment further comprises informing the subject that N-(l-methylpiperidi ⁇ -4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide ameliorates insomnia.
  • the informing comprises providing printed matter that N- (l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide ameliorates insomnia.
  • the printed matter is a label.
  • Another embodiment disclosed herein includes a method for decreasing the number of awakenings after sleep onset comprising administering N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpro ⁇ yloxy)phenylmethyl) carbamide to a subject in need of a decreased number of awakenings after sleep onset in an amount sufficient to decrease the number of awakenings after sleep onset.
  • Another embodiment disclosed herein includes a method for decreasing the time awake after sleep onset comprising administering N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide to a subject in need of decreased time awake after sleep onset in an amount sufficient to decrease time awake after sleep onset.
  • Another embodiment disclosed herein includes a method of manufacturing a pharmaceutical composition including obtaining a first dosage form comprising a first amount of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4- (2-methylpropyloxy)phenylmethyl) carbamide, obtaining a second dosage form comprising a second amount of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'- (4-(2-methylpropyloxy)phenylmethyl) carbamide, and packaging together the first dosage form and the second dosage form.
  • the first dosage form and the second dosage form each comprise an oral dosage form.
  • the first dosage form comprises a higher dose of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide than the second dosage form.
  • the amount of N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide is in the range of about 0.5 rag to about 50 mg. In one embodiment, the amount is in the range of about 1 mg to about 40 mg. In one embodiment, the amount is in the range of about 2.5 mg to about 30 mg. In one embodiment, the amount is in the range of about 5 mg to about 20 mg.
  • the administration does not affect a sleep parameter selected from the group consisting of sleep period time, total sleep time, sleep onset latency, number of stage shifts, total time awake, early morning wake, sleep efficiency index, microarousal index, and a REM sleep parameter.
  • the REM sleep parameter is selected from the group consisting of REM sleep duration, proportion of REM sleep, REM sleep latency, REM activity and REM density.
  • the amount results in a steady state blood plasma concentration of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the range of about 2 ng/mL to about 60 ng/mL. In one embodiment, the amount results in a steady state blood plasma concentration in the range of about 4 ng/mL to about 50 ng/mL. In one embodiment, the amount results in a steady state blood plasma concentration of in the range of about 6 ng/mL to about 40 ng/mL.
  • the amount results in a steady state blood plasma concentration in the range of about 8.5 ng/mL to about 35 ng/mL.
  • the subject is administered N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide once every day.
  • the subject is administered a first dosage form at least 24 hours prior to administration of a second dosage form.
  • the first dosage form comprises a higher dose than the second dosage form.
  • N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide is administered at a time other than immediately before a sleep period. In one embodiment, it is administered in the morning.
  • Another embodiment disclosed herein includes a packaged pharmaceutical composition, comprising N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in a container and instructions for using N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide to treat insomnia.
  • Another embodiment disclosed herein includes a packaged pharmaceutical composition, comprising N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in a container and instructions for using N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide to increase slow wave sleep, decrease the number of awakenings after sleep onset or decrease the time awake after sleep onset.
  • kits comprising a first dosage form of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy) ⁇ henylmethyl) carbamide and a second dosage form of N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide.
  • the first dosage form contains a higher dose of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide than the second dosage form.
  • the kit further comprises instructions for taking the first dosage form at least 24 hours before taking the second dosage form.
  • the instructions are on a label.
  • the instructions further inform a subject that N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide increases slow wave sleep.
  • the instructions further inform a subject that N-(l-methylpiperidin-4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide ameliorates insomnia. In one embodiment, the instructions further inform a subject that N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide decreases the number of awakenings after sleep onset.
  • the instructions further inform a subject that N-(I- methylpiperidin-4-yl)-N-(4-f ⁇ uorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide decreases the time awake after sleep onset.
  • FIGURE 1 is a graph depicting the change in slow wave sleep duration from baseline for placebo and various doses of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide.
  • FIGURE 2 is a graph depicting the correlation between slow wave sleep duration and blood plasma level of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N ' -(4-(2-methylpropyloxy)phenylmethyl) carbamide.
  • FIGURE 3 is a graph depicting the change in number of awakenings from baseline for placebo and various doses of N-(l-methyl ⁇ iperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide.
  • the present application relates to the use of compounds that are inverse agonists or antagonists of a serotonin receptor to treat a sleep disorder.
  • the compounds are inverse agonists or antagonists of a 5-HT2A receptor.
  • the compounds are selective inverse agonists or antagonists.
  • the sleep disorder is insomnia.
  • the insomnia is sleep maintenance insomnia.
  • the sleep maintenance insomnia is caused by other disorders.
  • the sleep maintenance insomnia is comorbid with a psychiatric disorder including, but not limited to, schizophrenia, affective disorders including MDD, bipolar 2 depression, bipolar 1 mania, rapid cyclers, dysthymia, PTSD (e.g., nightmares or arousals), alcoholism, substance abuse, drug withdrawal, and anxiety.
  • a psychiatric disorder including, but not limited to, schizophrenia, affective disorders including MDD, bipolar 2 depression, bipolar 1 mania, rapid cyclers, dysthymia, PTSD (e.g., nightmares or arousals), alcoholism, substance abuse, drug withdrawal, and anxiety.
  • the compounds described herein may be administered in combination with an SSRI or NASR to decrease insomnia, increase efficacy, decrease sexual side effects and akathisia, provide a faster response, and limit treatment resistance.
  • the sleep maintenance insomnia is caused by a neurological disorder, including but not limited to Parkinson's disease, multisystems atrophy, migraines, multiple sclerosis, Huntington's Chorea, "Sun-downing" in DAT and other dementia's, and epilepsy.
  • a neurological disorder including but not limited to Parkinson's disease, multisystems atrophy, migraines, multiple sclerosis, Huntington's Chorea, "Sun-downing" in DAT and other dementia's, and epilepsy.
  • Other disorders that may cause sleep maintenance insomnia that can be treated using the compounds described herein include, but are not limited to, rheumatoid and osteo-arthritis and other chronic disorders with pain, fibro-myalgia, and female menopause.
  • the compounds described herein are administered to a patient to increase slow wave sleep in the patient for any purpose.
  • the compounds decrease the number of awakenings after sleep onset and the time awake after sleep onset.
  • the compounds described herein are administered to a patient to treat other sleep related disorders including periodic limb movement syndrome or obstructive sleep apnea.
  • the compounds achieve the desired effect on slow wave sleep, number of awakenings after sleep onset, and the time awake after sleep onset without affecting other sleep parameters.
  • sleep parameters unaffected include sleep period time, total sleep time, sleep onset latency, number of sleep stage shifts, total time awake, early morning wake, sleep efficiency index, microarousal index, and REM sleep parameters (e.g., REM sleep duration, proportion of REM sleep, REM sleep latency, REM activity, and REM density).
  • the compounds are administered at a dose between about 0.5 mg and about 50 mg, between about 1 mg and about 40 mg, between about 2.5 mg and about 30 mg, or between about 1 mg and about 20 mg.
  • the dosage and administration cyst are sufficient to achieve a steady state blood plasma concentration of between about 2 ng/ml and about 60 ng/ml, between about 4 ng/ml and about 50 ng/ml, between about 6 ng/ml and about 40 ng/ml, or between about 8.5 ng/ml and about 35 ng/ml.
  • the dosage is varied over the dosage regimen.
  • the first administration or series of administrations have a higher dosage than subsequent administrations.
  • the compound administered has a relatively high half-life such that after an initial dosage sufficient to raise the blood plasma concentration to a desired level, subsequent dosages sufficient to maintain a desired steady state blood plasma concentration can be lower.
  • pharmaceutical compositions comprising multiple doses are packaged together.
  • a sleep-inducing agent is administered in combination with the compounds described herein.
  • the sleep-inducing agent may be administered to induce onset of sleep in the patient while the inverse agonist or antagonist of a serotonin receptor may be administered to maintain sleep in the patient.
  • the compounds described herein may be administered to maintain slow wave sleep in the patient.
  • suitable sleep-inducing agents include AMBIEN®, indiplon, LUNESTA®, and melatonin.
  • administration in “combination” it is meant that the two or more agents may be found in the patient's bloodstream at the same time, regardless of when or how they are actually administered.
  • the agents are administered simultaneously.
  • administration in combination is accomplished by combining the agents in a single dosage form.
  • the agents are administered sequentially.
  • the agents are administered through the same route, such as orally.
  • the agents are administered through different routes, such as one being administered orally and another being administered intravenously.
  • the pharmacokinetics of the two or more agents are substantially the same.
  • the compounds described herein are long acting (e.g., have a long half-life), allowing them to be given relatively infrequently and at times other than immediately prior to a desired sleep period.
  • the compounds have a half-life long enough so that that they don't need to be administered more often than once a day.
  • the compound may be administered once every 1, 2, 3, 4, or 5 days and still provide sleep maintenance properties during each sleep period.
  • the half-life of the compound is from about 10 hours to about 100 hours, from about 20 hours to about 60 hours, or from about 35 hours to about 55 hours. In some embodiments, the compound is not administered immediately prior to a sleep period.
  • the compound may be administered in the morning or afternoon. In one embodiment, the compound is administered in the morning.
  • the long acting properties of the compounds eliminate or reduce any withdrawal effects experienced by patients as either a co-administered sleep-inducing agent or the inverse agonist or antagonist of a serotonin receptor wears off. For example, many short acting sleep agents cause a patient to wake in the middle of their sleep period and experience a withdrawal effect. In some embodiments, patients taking a compound described herein experience no such withdrawal effect. Definitions
  • constitutive activity is defined as the basal activity of a receptor which is independent of the presence of an agonist. Constitutive activity of a receptor may be measured using a number of different methods, including cellular (e.g., membrane) preparations (see, e.g., Barr &. Manning, J. Biol. Chem.
  • agonist is defined as a compound that increases the activity of a receptor when it contacts the receptor.
  • antagonist is defined as a compound that competes with an agonist or inverse agonist for binding to a receptor, thereby blocking the action of an agonist or inverse agonist on the receptor.
  • an antagonist also known as a “neutral” antagonist
  • inverse agonist is defined as a compound that decreases the basal activity of a receptor (i.e., signaling mediated by the receptor). Such compounds are also known as negative antagonists.
  • An inverse agonist is a ligand for a receptor that causes the receptor to adopt an inactive state relative to a basal state occurring in the absence of any ligand.
  • an antagonist can inhibit the activity of an agonist
  • an inverse agonist is a ligand that can alter the conformation of the receptor in the absence of an agonist.
  • Bond et al. in Nature 374:272 (1995). More specifically, Bond et al.
  • an inverse agonist can additionally manifest its activity in the absence of an agonist by inhibiting the spontaneous conversion of an unliganded receptor to an active conformation.
  • 5-HT2A receptor is defined as a receptor, having an activity corresponding to the activity of the human serotonin receptor subtype, which was characterized through molecular cloning and pharmacology as detailed in Saltzman et al., Biochem. Biophys. Res. Comm. 181:1469-78; and Julius et al., Proc. Natl. Acad. ScL USA 87:928-932.
  • subject refers to an animal, preferably a mammal, most preferably a human, who is the object of treatment, observation or experiment.
  • selective is defined as a property of a compound whereby an amount of the compound sufficient to effect a desired response from a particular receptor type, subtype, class or subclass causes a substantially smaller or no effect upon the activity other receptor types.
  • the terms "selectivity" or “selective,” in relation to an inverse agonist, are understood as a property of a compound of the invention whereby an amount of compound that effectively inversely agonizes the 5 -HT2 A. receptor, and thereby decreases its activity, causes little or no inverse agonistic or antagonistic activity at other, related or unrelated, receptors.
  • certain compounds of the invention have been found not to interact strongly with other serotonin receptors (S-HT IA, IB, ID, IE, IF, 2B, 2C 5 4 A, 6, and 7) at concentrations where the signaling of the 5-HT2 A receptor is strongly or completely inhibited.
  • the compounds of the invention are also selective with respect to other monoamine-binding receptors, such as the dopaminergic, histaminergic, adrenergic and muscarinic receptors.
  • monoamine-binding receptors such as the dopaminergic, histaminergic, adrenergic and muscarinic receptors.
  • Compounds that are highly selective for 5-HT2A receptors may have a beneficial effect in the treatment of psychosis, schizophrenia or similar neuropsychiatric disorders, while avoiding adverse effects associated with drugs hitherto suggested for this purpose.
  • the EC50 for an agonist is intended to denote the concentration of a compound needed to achieve 50% of a maximal response seen in R-SAT.
  • EC50 is intended to denote the concentration of a compound needed to achieve 50% inhibition of an R-SAT response from basal, no compound, levels.
  • aryl is intended to mean a carbocyclic aromatic ring or ring system. Moreover, the term “aryl” includes fused ring systems wherein at least two aryl rings, or at least one aryl and at least one C 3 - 8 -cycloalkyl share at least one chemical bond. Some examples of “aryl” rings include optionally substituted phenyl, naphthalenyl, phenanthrenyl, anthracenyl, tetralinyl, fluorenyl, indenyl, and indanyl.
  • aryl relates to aromatic, preferably benzenoid groups, connected via one of the ring-forming carbon atoms, and optionally carrying one or more substituents selected from heterocyclyl, heteroaryl, halo, hydroxy, amino, cyano, nitro, alkylamido, acyl, Ci- 6 alkoxy, Ci -6 alkyl, Ci- 6 hydroxyalkyl, Ci -6 aminoalkyl, Ci-e alkylamino, alkylsulfenyl, alkylsulf ⁇ nyl, alkylsulfonyl, sulfamoyl, or trifluoromethyl.
  • the aryl group may be substituted at the para and/or meta positions.
  • aryl groups include, but are not limited to, phenyl, 3-halophenyl, 4-halophenyl, 3- hydroxyphenyl, 4-hydroxyphenyl, 3-aminophenyl, 4-aminophenyl, 3-methylphenyl, 4- methylphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 4-trifluoromethoxyphenyl 3- cyanophenyl, 4-cyanophenyl, dimethylphenyl, naphthyl, hydroxynaphthyl, hydroxymethylphenyl, trifluoromethylphenyl, alkoxyphenyl, 4-mo ⁇ holin-4-ylphenyl, 4- pyrrolidin-1-ylphenyl, 4-pyrazolylphenyl, 4-triazolylphenyl, and 4-(2-oxopyrrolidin-l- yl)phenyl.
  • heteroaryl is intended to mean a heterocyclic aromatic group where one or more carbon atoms in an aromatic ring have been replaced with one or more heteroatoms selected from the group comprising nitrogen, sulfur, phosphorous, and oxygen.
  • heteroaryl comprises fused ring systems wherein at least one aryl ring and at least one heteroaryl ring, at least two heteroaryl rings, at least one heteroaryl ring and at least one heterocyclyl ring, or at least one heteroaryl ring and at least one C 3 .s-cycloalkyl ring share at least one chemical bond.
  • heteroaryl is understood to relate to aromatic, C 3 - 8 cyclic groups further containing one oxygen or sulfur atom or up to four nitrogen atoms, or a combination of one oxygen or sulfur atom with up to two nitrogen atoms, and their substituted as well as benzo- and pyrido-fused derivatives, preferably connected via one of the ring-forming carbon atoms.
  • Heteroaryl groups may carry one or more substituents, selected from halo, hydroxy, amino, cyano, nitro, alkylamido, acyl, Ci-6-alkoxy, C1-6- alkyl, Cj-6-hydroxyalkyl, Ci -6 -aminoalkyl, Ci.
  • heteroaryl groups may be five- and six-membered aromatic heterocyclic systems carrying 0, 1, or 2 substituents, which may be the same as or different from one another, selected from the list above.
  • heteroaryl groups include, but are not limited to, unsubstituted and mono- or di-substituted derivatives of furan, benzofuran, thiophene, benzothiophene, pyrrole, pyridine, indole, oxazole, benzoxazole, isoxazole, benzisoxazole, thiazole, benzothiazole, isothiazole, imidazole, benzimidazole, pyrazole, indazole, tetrazole, quionoline, isoquinoline, pyridazine, pyrimidine, purine and pyrazine, which are all preferred, as well as furazan, 1,2,3-oxadiazole, 1,2,3-thiadiazole, 1,2,4- thiadiazole, triazole, benzotriazole, pteridine, phenoxazole, oxadiazole, benzopyrazole, quinolizine,
  • the substituents are halo, hydroxy, cyano, O-Ci- 6 -alkyl, Ci ⁇ -alkyl, hydroxy-Ci- ⁇ -alkyl, amino-Ci. 6 -al.cyl.
  • alkyl d- 6 -alkyl
  • alkyl d- 6 -alkyl
  • d- 6 -alkyl are intended to mean a linear or branched saturated hydrocarbon chain wherein the longest chain has from one to six carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec- butyl, tert-butyl, pentyl, isopentyl, neopentyl, and hexyl.
  • An alkyl chain may be optionally substituted.
  • “Lower alkyl groups” are Ci -6 cyclic, straight-chained or branched aliphatic substituent groups connected via a carbon atom. Examples include methyl, ethyl, propyl, butyl, methylbutyl, cyclopropyl, cyclohexyl, iso-propyl, tert-butyl.
  • C 2 - 8 -alkenyl is intended to mean a linear or branched hydrocarbon group having from two to eight carbon atoms and containing one or more double bonds.
  • Some examples of C 2 - 8 -alkenyl groups include allyl, homo-allyl, vinyl, crotyl, butenyl, pentenyl, hexenyl, heptenyl and octenyl.
  • C2- 8 -alkenyl groups with more than one double bond include butadienyl, pentadienyl, hexadienyl, heptadienyl, heptatrienyl and octatrienyl groups as well as branched forms of these.
  • the position of the unsaturation (the double bond) may be at any position along the carbon chain.
  • C 2 - 8 -alkynyl is intended to mean a linear or branched hydrocarbon group containing from two to eight carbon atoms and containing one or more triple bonds.
  • Some examples of C 2 - 8 -alkynyl groups include ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl and octynyl groups as well as branched forms of these.
  • the position of unsaturation (the triple bond) may be at any position along the carbon chain. More than one bond may be unsaturated such that the "C2-8-alkynyl" is a di-yne or enedi-yne as is known to the person skilled in the art.
  • Cs-s-cycloalkyl is intended to cover three-, four-, five-, six-, seven-, and eight-membered rings comprising carbon atoms only.
  • a Ca- ⁇ -cycloalkyl may optionally contain one or more unsaturated bonds situated in such a way, however, that an aromatic ⁇ -electron system does not arise.
  • Ca ⁇ -cycloalkyl are the carbocycles cyclopropane, cyclobutane, cyclopentane, cyclopentene, cyclopentadiene, cyclohexane, cyclohexene, 1,3-cyclohexadiene, 1,4-cyclohexadiene, cycloheptane, cycloheptene.
  • Cyclic organyl groups are aliphatic, alicyclic groups in which carbon atoms form a ring. In preferred embodiments containing three, four, five, six or seven carbon atoms, the ring, as a substituent, is connected either directly via one of the ring atoms or via one or more appended carbon atoms. Particular examples of such groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, cyclohexylethyl groups, and the like.
  • Straight-chained acyclic organyl groups are substituent groups consisting of a linear arrangement of carbon atoms, where accordingly each carbon atom binds a maximum of two other carbon atoms, connected through single, double, or triple bonds.
  • the straight-chained organyl groups may contain none, one, or several multiple bonds, and are, for example, commonly referred to as alkyl, alkenyl or alkynyl, or alkadienyl groups, respectively.
  • straight-chained organyl groups include methyl, ethyl, propyl, butyl, pentyl, hexyl, propenyl, butenyl, pentadienyl, propargyl, butynyl.
  • Branched acyclic organyl groups are substituent groups consisting of a branched arrangement of carbon atoms, where accordingly one or more carbon atoms may bind more than two other carbon atoms, connected through single, double, or triple bonds.
  • the branched organyl groups may contain none, one, or several multiple bonds. Examples of branched organyl groups include iso-propyl, iso-butyl, tert-butyl, methylbutyl, methylbutenyl, methylbutynyl.
  • C)- 6 alkoxy and "lower alkoxy groups” are understood as C ] . 6 cyclic or acyclic organyl groups connected, as substituents, via an oxygen atom. Examples of lower alkoxy groups include methoxy, ethoxy, iso-propoxy, butoxy, tert- butoxy, cyclopropyl, cyclobutyl, cyclopropylmethyl, and cyclobutylmethyl.
  • Ci. 6 alkylamino and “lower alkylamino groups” are understood as lower alkyl groups connected, as substituents, via a nitrogen atom, which may carry one or two lower alkyl groups. Particular examples include methylamino, dimethylamino, iso-propylamino. Optionally, lower aminoalkyl groups may consist of 4-6 membered nitrogen-containing rings, such as pyrrolidino.
  • aminoalkyl and “lower aminoalkyl groups” are understood as lower alkyl groups carrying, as a substituent, an additional amino group. Examples include aminomethyl and aminoethyl.
  • Ci- 6 -hydroxyalkyl and “lower hydroxyalkyl groups” are understood as lower alkyl groups carrying, as a substituent, an additional hydroxy group. Examples include hydroxymethyl, hydroxyethyl, 2-hydroxy-2-propyl, hydroxypentyl.
  • heterocyclyl is intended to mean three-, four-, five-, six-, seven-, and eight-membered rings wherein carbon atoms together with from 1 to 3 heteroatoms constitute the ring.
  • a heterocyclyl may optionally contain one or more unsaturated bonds situated in such a way, however, that an aromatic ⁇ -electron system does not arise.
  • the heteroatoms are independently selected from oxygen, sulfur, and nitrogen.
  • a heterocyclyl may further contain one or more carbonyl or thiocarbonyl functionalities, so as to make the definition include oxo-systems and thio- systems such as lactams, lactones, cyclic imides, cyclic thioimides, cyclic carbamates, and the like.
  • Heterocyclyl rings may optionally also be fused to aryl rings, such that the definition includes bicyclic structures.
  • Preferred such fused heterocyclyl groups share one bond with an optionally substituted benzene ring.
  • benzo-fused heterocyclyl groups include, but are not limited to, benzimidazolidinone, tetrahydroquinoline, and methylenedioxybenzene ring structures.
  • heterocyclyls include, but are not limited to, tetrahydrothiopyran, 4H-pyran, tetrahydropyran, piperidine, 1,3-dioxin, 1,3-dioxane, 1,4- dioxin, 1,4-dioxane, piperazine, 1,3-oxathiane, 1 ,4-oxathiin, 1,4-oxathiane, tetrahydro- 1 ,4-thiazine, 2//-l,2-oxazine , maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, morpholine, trioxane, hexahydro-1,3,5- triazine, tetrahydrothiophene, tetrahydrofuran, pyrroline, pyrrolidine,
  • Binding to the heterocycle may be at the position of a heteroatom or via a carbon atom of the heterocycle, or, for benzo-fused derivatives, via a carbon of the benzenoid ring.
  • the term "(heterocyclyl)Ci-6-alkyl” is understood as heterocyclyl groups connected, as substituents, via a lower alkylene, each as defined herein.
  • the heterocyclyl groups of (heterocyclyl)Ci- 6 -alkyl groups may be substituted or unsubstituted.
  • (aryl)Ci- 6 -alkyl” and “aralkyl” are intended to mean an aryl group connected, as a substituent, via a lower alkylene, each as defined herein.
  • the aryl groups of (ary ⁇ Ci- ⁇ -alkyl may be substituted or unsubstituted. Examples include benzyl, substituted benzyl, 2-phenylethyl, 3-phenylpropyl, and naphthylalkyl.
  • heteroarylCi. 6 -alkyl and “heteroaralkyl” are understood as heteroaryl groups connected, as substituents, via a lower alkylene, each as defined herein.
  • the heteroaryl groups of heteroaralkyl groups may be substituted or unsubstituted. Examples include 2-thienylmethyl, 3-thienylmethyl, furylmethyl, thienylethyl, pyrrolylalkyl, pyridylalkyl, isoxazolylalkyl, imidazolylalkyl, and their substituted as well as benzo-fused analogs.
  • (cyclo alky I)C i- ⁇ -alkyl is intended to mean a cycloalkyl groups connected, as substituents, via a lower alkylene, each as defined herein.
  • O-Ci-e-alkyl is intended to mean Ci-g- alkyloxy, or alkoxy, such as methoxy. ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, pentyloxy, isopentyloxy, neopentyloxy and hexyloxy.
  • the definition of "O-Ci. 6 -alkyl” is intended to cover cyclic alkoxy groups having a maximum of six carbon atoms. Illustrative non-limiting examples of cyclic alkoxy groups include cyclobutyloxy, cyclopropylmethyloxy, cyclohexyloxy, and the like.
  • lower alkylene means a bivalent hydrocarbon tether, containing from one to six carbon atoms. Additionally, “lower alkylene” tethers may optionally contain one or more substituents selected from Ci- ⁇ alkyl, halogen, hydroxyl, and amino. Non-limiting examples of “lower alkylene” groups are methylene, ethylene, propylene, tetramethylene, hexamethylene.
  • Vinylene groups are ethene-l,2-diyl groups (— CHCH-) having (E) or (Z) configuration.
  • Acyl groups are hydrogen or lower alkyl groups connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl.
  • haloalkyl hydroxyalkyl
  • aminoalkyl are intended to cover Cj-6-alkyl groups, defined above, carrying at least one halogen, hydroxy group, or amino group, respectively.
  • halogen includes fluorine, chlorine, bromine and iodine.
  • the term "optionally substituted” is intended to mean that the group in question may be substituted one or several times, such as 1 to 5 times, or 1 to 3 times, or 1 to 2 times, with one or more groups selected from Ci-e-alkyl, Ci- 6 -alkoxy, oxo (which may be represented in the tautomeric enol form), carboxyl, amino, hydroxy (which when present in an enol system may be represented in the tautomeric keto form), nitro, alkylsulfonyl, alkylsulfenyl, alkylsulf ⁇ nyl,Ci- 6 -alkoxycarbonyl, d- ⁇ -alkylcarbonyl, forrnyl, amino, mono- and di(Ci- 6 -alkyl)amino; carbamoyl, mono- and di(C]- 6 - alkyl)aminocarbonyl
  • salts is intended to mean pharmaceutically acceptable acid addition salts obtainable by treating the base form of a functional group, such as an amine, with appropriate acids such as inorganic acids, for example hydrohalic acids; typically hydrochloric, hydrobromic, hydrofluoric, or hydroiodic acid; sulfuric acid; nitric acid; phosphoric acid and the like; or organic acids, for example acetic, propionic, hydroacetic, 2-hydroxypropanoic acid, 2-oxopropanoic acid, ethandioic, propanediol, butanedioic, (Z)-2-butenedioic, (E)-butenedioic, 2-hydroxybutanedioic, 2,3-dihydroxybutanedioic, 2- hydroxy-l,2,3-propanetricarboxylic, methanesulfonic, ethanesulfonic, benzenesulfonic, 4- methylbenzenesulfonic
  • the present invention includes within its scope prodrugs of the compounds of this invention.
  • prodrugs are inactive derivatives of the compounds of this invention that are readily convertible in vivo into the required compound.
  • Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Design of Prodrugs, (ed. H. Bundgaard, Elsevier, 1985). Metabolites of these compounds include active species that are produced upon introduction of compounds of this invention into the biological milieu.
  • the compounds according to the invention may exist as a racemate or as enantiomers. It should be noted that all such isomers and mixtures thereof are included in the scope of the present invention. Furthermore, some of the crystalline forms for compounds of the present invention may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds of the present invention may form solvates with water (i.e., hydrates) or common organic solvents. Such solvates are also included in the scope of this invention.
  • Such isomers may be separated by conventional techniques such as preparative chiral chromatography.
  • the compounds may be prepared in racemic form or individual enantiomers may be prepared by stereoselective synthesis or by resolution.
  • the compounds may be resolved into their component enantiomers by standard techniques, such as the formation of diastereomeric pairs by salt formation with an optically active acid, such as (-)-di-p-toIuoyl-d-tartaric acid and/or (+)- di-p-toluoyl-1-tartaric acid, followed by fractional crystallization and regeneration of the free base.
  • the compounds may also be resolved using a chiral auxiliary by formation of diastereomeric derivatives such as esters, amides or ketals followed by chromatographic separation and removal of the chiral auxiliary.
  • One embodiment includes the use of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide, a 5-HT2A inverse agonist which has the structure:
  • R is a hydrogen, a cyclic or straight-chained or branched acyclic organyl group, a lower hydroxyalkyl group, a lower aminoalkyl group, or an aralkyl or heteroaralkyl group; and n can be 0, 1, or 2.
  • X] is methylene, vinylene, or an NH or N(lower alkyl) group
  • X 2 is methylene , or, when X] is methylene or vinylene, X 2 is methylene or a bond; or when Xj is methylene, X 2 is O, S, NH, or N(lower alkyl) or a bond.
  • Y] is methylene and Y 2 is methylene, vinylene, ethylene, propylene, or a bond; or Yj is a bond and Y2 is vinylene; or Yj is ethylene and Y 2 is O, S, NH, or N(lower alkyl).
  • Ari and Ar 2 each independently is unsubstituted or substituted aryl or heteroaryl groups.
  • W is oxygen or sulfur.
  • Yi is methylene and Y 2 is a bond, methylene, ethylene, or vinylene; or Yi is ethylene and Y2 is O or S; and Xj is methylene and X 2 is a bond, methylene, O, or S; or Xi is NH or N(lower alkyl) and X 2 is methylene.
  • Z is
  • Arj and Ar 2 independently are mono- or disubstituted phenyl groups.
  • the R of Formula (I) is a hydrogen, a lower alkyl group, a cyclic organyl group, or a substituted or unsubstituted aralkyl or heteroaralkyl group; n is 1; Yi is methylene, and Y 2 is a bond, methylene, ethylene, or vinylene; Xi is methylene and X2 is a bond, or Xi is NH or N(lower alkyl) and X 2 is methylene; andAri and Ar 2 are phenyl groups, independently / ⁇ -substituted with groups selected from lower alkyl, lower alkoxy and halogen.
  • R N is hydrogen, lower alkyl, aralkyl, or heteroaralkyl
  • Ar L is selected from lower alkyl, lower alkoxy and halogen
  • Ar R is selected from lower alkyl, lower alkoxy and halogen
  • k is 1 or 2
  • a " is a suitable anion.
  • Forma I refers to compounds of Formulae (I) and (Ia).
  • Suitable embodiments of the compounds of Formula I may be selected from the group consisting, of: iV-((4-methylphenyl)methyl)-iV " -(piperidin-4-yl)-7V- phenylmethylcarbamide;
  • 2-(4-ethoxyphenyl)-iV -(4-fluoroben2yl)-//-( 1 -methylpiperidin-4-yl) acetamide; 2-(4-Chlorophenyl)-N-(4-methylbenzyl)-N-(l-isopropylpiperidin-4-yl)- acetamide;
  • the compound of Formula I is selected from the group consisting of:
  • the compound of Formula I is selected from the group consisting of:
  • the compound of Formula I is 2-(4- methoxyphenyl)-N-(4-methylbenzyl)-N-(8-methyl-8-aza-bicyclo[3.2.1 ]oct-3-yl)- acetamide.
  • the compound of Formula I is selected from the group consisting of:
  • the compound of Formula I is selected from the group consisting of:
  • the compound of Formula I is 2-(4- Methoxyphenyl)-N-(4-methylbenzyl)-N-(8-methyl-8-aza-bicyclo[3.2.1]octen-3-yl)- acetamide.
  • R 1 is selected from the group consisting of optionally substituted heterocyclyl, and optionally substituted (heterocyclyl)Ci-6-alkyl.
  • R 2 and R 3 are independently selected from the group consisting of hydrogen, Ci-6-alkyl and halogen or such that R 2 together with R 3 forms a ring.
  • R 2 and R 3 together can form a 3-, A-, S-, 6-, or 7-membered ring system with the atoms of the piperidine ring.
  • m 1 is selected from the group consisting of 0, 1 , and 2.
  • n 1 is selected from the group consisting of 1, 2, and 3;
  • a ⁇ 3 is an optionally substituted aryl or heteroaryl.
  • the aryl or heteroaryl can be optionally substituted with a substituent selected from the group consisting of Ci- ⁇ -alkyl, Ci ⁇ -alkoxy, carboxyl, amino, hydroxy, thiol, nitro, cyano, guanidino, carbamido and halogen.
  • Wi is selected from the group consisting of O and S.
  • X 3 is selected from the group consisting of optionally substituted methylene, optionally substituted ethylene, optionally substituted propylene, optionally substituted vinylene, and CH 2 N(R N1 ), wherein R N1 is selected from hydrogen and Cj-e-alkyl.
  • a ⁇ 4 is an optionally substituted aryl or heteroaryl.
  • the heterocyclyl or (heterocyclyl)C]_ 6 -alkyl of R 1 may be optionally substituted.
  • the substituent may be selected from halogen, hydroxy, alkyl, alkoxy, and amino.
  • the substituent may be on the alkyl chain or the ring system. In further embodiments the substituent is on the ring system.
  • the heterocyclyl ring in R 1 may be selected from the group consisting of tetrahydrothiopyran, 4H-pyran, tetrahydropyran, piperidine, 1,3-dioxin, 1,3-dioxane, 1,4-dioxin, 1,4-dioxane, piperazine, 1,3-oxathiane, 1 ,4-oxathiin, 1,4-oxathiane, tetrahydro-l,4-thiazine, 2H-l,2-oxazine, maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, morpholine, trioxane, hexahydro-l,3,5-triazine, tetrahydrothiophene, tetrahydrofuran, pyrroline, pyrrol
  • the azacyclic ring may be a 5, 6, or 7-membered ring as reflected in that m' may be selected from 0, 1 and 2. In certain embodiments, however, the azacyclic ring is a 6-membered ring, wherein m' is 1.
  • the azacyclic ring may be substituted with R 2 and R 3 .
  • R 2 and R 3 may be independently selected from the group consisting of hydrogen, Ci- 6 -alkyl, and halogen, or such that R together with R forms a ring. That is to say that R 2 and R 3 may be biradicals which combine to form a 3-, 4-, 5-, 6-, or 7-membered ring system with the atoms of the azacyclic ring.
  • the azacyclic ring system is selected from wherein R 7 and R 8 are independently selected from the group consisting of hydrogen, halogen, hydroxyl, and Cj. ⁇ alkyl. In certain embodiments R 7 and R 8 are hydrogen.
  • R 2 and R 3 are hydrogen.
  • R 1 is an optionally substituted (heterocyclyl)Ci_ 6-alkyl. In certain of these embodiments, R 1 is an optionally substituted (heterocyclyl)methyl, an optionally substituted (heterocyclyl)ethyl, or an optionally substituted (heterocyclyl)propyl. In other embodiments, R 1 is an optionally substituted (hetero cycly l)ethyl .
  • Ar 3 is linked to a central nitrogen atom via a short aliphatic chain 1, 2, or 3 carbon atoms in length.
  • n' is 1, resulting in a methylene spacer between the central nitrogen atom and Ar 3 .
  • Ar 3 may be an optionally substituted aryl or heteroaryl.
  • Ar 3 is an optionally substituted aryl.
  • the central nitrogen atom is linked to an optionally substituted benzyl group.
  • Ar 3 is an optionally substituted aryl, which may be a 4-substituted aryl.
  • the 4-substituent of the 4-substituted aryl may be any substituent known to the person skilled in the art, such as a C 1 - 6 -alkyl, Q- 6 -alkoxy, carboxyl, amino, hydroxy, thiol, nitro, cyano, guanidino, carbamido and halogen.
  • the halogen is fluoro, while in other embodiments, the halogen is chloro.
  • Ar 3 is selected from the group consisting of alkyl-substituted phenyl, alkoxy-substituted phenyl, halogen-substituted phenyl, hydroxy- substituted phenyl and amino-substituted phenyl.
  • the substituent may be present 0 to 5 times, or 0 to 4 times, or 0 to 3 times, such as 0, 1 , 2, or 3 times. In certain embodiments, the substituent is present 1 to 2 times.
  • Ar 3 is a 4-substituted aryl selected from the group consisting of 4-halophenyl and 4-alkylphenyl.
  • the phenyl group is 4-fluorophenyl.
  • Ar 3 is an optionally substituted heteroaryl.
  • the heteroaryl may be substituted with substituents known to the person skilled in the art, such as a Ci-g-alkyl, C]_6-alkoxy, carboxyl, amino, hydroxy, thiol, nitro, cyano, guanidino, carbamido and halogen.
  • the central nitrogen is linked to Ar4 via a 2 to 4 carbon spacer unit.
  • This spacer unit comprises a carbonyl or thiocarbonyl function wherein Wi is selected from the group consisting of oxygen and sulfur. In some embodiments Wi is oxygen.
  • X3 may be selected from the group consisting of optionally substituted methylene, optionally substituted ethylene, optionally substituted propylene, optionally substituted vinylene, and CH 2 N(R 1 "").
  • X 3 may extend the spacer unit by 1 to 3 atoms between the central nitrogen and Ar 4 and render the central nitrogen part of an amide or carbamide.
  • X 3 is selected from the group consisting of optionally substituted methylene, optionally substituted ethylene, and CH 2 N(R 1 " 11 ).
  • X 3 is an optionally substituted methylene, or CH 2 N(R N1 ), wherein R Nl may be hydrogen.
  • Ar 4 may be an optionally substituted aryl or heteroaryl. In certain embodiments, Ar 4 is an optionally substituted aryl. In some embodiments, Ai4 is a 4-substituted aryl.
  • Ar 4 may be selected from the group consisting of alkoxy-substituted phenyl, halogen-substituted phenyl, hydroxy-substituted phenyl, amino-substituted phenyl, and heterocyclyl-substituted phenyl.
  • Ar 4 is a 4-substituted aryl wherein the substituent is selected from the group consisting of alkyl, alkoxy, halogen, hydroxy, amino, alkylamino, heterocyclyl, and heteroaryl.
  • the substituent on Ar 4 is selected from chloro, fluoro, hydroxy, methoxy, ethoxy, propoxy, isopropoxy, n- butoxy, sec-butoxy, tert-butoxy, trifluoromethoxy, N-morpholinyl, N-pyrrolidinyl, N- pyrazolyl, N-triazolyl and 2-oxopyrrolidinyl.
  • Suitable embodiments of the compounds of Formula II can be selected from the group consisting of: iV- ⁇ l-[2-(l,3-Dioxolan-2-yl)ethyl]piperidin-4-yl ⁇ -N-(4-fluorobenzyl)-N'-
  • X4 is selected from the group consisting of CH2, CH 2 CH 2 , CH 2 O, OCH 2 , O, CH 2 S 5 SCH 2 , S, CH 2 N(R N2 ), N(R N2 )CH 2 and N(R N2 ); wherein R N2 is selected from hydrogen and C 1 . 6 alkyl.
  • W 2 is selected from the group consisting of O and S.
  • Z 3 is absent or selected from the group consisting of CH and N.
  • R 9 is hydrogen, or an optionally substituted substituent selected from the group consisting of Q-e alkyl, C 2 - 8 alkenyl, C 2 -S alkynyl, C3.8 cycloalkyl, aryl, heteroaryl, aryl(Ci ⁇ alkyl), heteroaryl(Ci. 6 alkyl), heterocyclylCCj- ⁇ alkyl), C3.8 cyc!oalkyl(Ci. 6 alkyl), hydroxy(C]. 6 alkyl), amino(Ci. 6 alkyl), and halo(C 1 . 6 alkyl).
  • m is selected from the group consisting of 0 and 1.
  • R 12 , R 13 , and R 14 are independently hydrogen, or an optionally substituted substituent selected from the group consisting of Ci -6 alkyl, aryl(C
  • R 10 and R 11 are independently selected from the group consisting of hydrogen, halogen, hydroxy, and optionally substituted C 1. 6 alkyl or selected such that R 10 and R 1 ' together form a ring system such that is selected from the group consisting of
  • R 15 and R 16 are independently selected from the group consisting of hydrogen, halogen, hydroxy, and Cj. 6 alkyl.
  • R 12 , R 13 , and R 14 may be independently selected from the group consisting of 4-monosubstituted-aryl(Ci -6 alkyl), and 4- monosubstituted-heteroaryl(Ci.6 alkyl).
  • the other of the at least two of R 12 , R 13 , and R 14 independently selected from the group consisting of aryl(Ci- 6 alkyl) and heteroaryl(Ci_6 alkyl) is selected from the group consisting of (O-Ci-6 alkyl)-substituted- aryl(C i-6 alkyl), and (O-Ci ⁇ alkyl)-substituted-heteroaryl(Ci-6 alkyl).
  • At least one of R 12 , R 13 , and R 14 is selected from the group consisting of fluoro-substituted-aryl(Ci- 6 alkyl), and fluoro-substituted- heteroaryl(Ci-6 alkyl).
  • R 12 , R 13 , and R 14 are independently selected from the group consisting of aryl(Ci-6 alkyl), heteroaryl(Ci- 6 alkyl), and heterocycIyl(Ci-6 alkyl) are each substituted 1, 2, or 3 times, with a substituent selected from the group consisting of halogen and optionally substituted O-C L ⁇ -alkyl.
  • the halogen is fluorine.
  • -6 alkyl), heterocyclyl(Ci_6 alkyl) is substituted 1 to 3 times, such as 1, 2, or 3 times with an optionally substituted O-Ci -6 -alkyl, such as a fluorinated O-Ci ⁇ -alkyl.
  • At least two of R 12 , R I3 5 and R 14 are optionally substituted aryl(Ci. 6 alkyl). In a preferred embodiment, at least two of R 12 , R 13 , and R 14 are optionally substituted benzyl.
  • R 12 , R 13 , and R 14 are independently selected from the group consisting of aryl(Ci- 6 alkyl), heteroary ⁇ C ⁇ alkyl), heterocyclyl(Ci. 6 alkyl).
  • the d. 6 alkyl of the aryl(Ci_ 6 alkyl), heteroaryl(C 1-6 alkyl), heterocyclyl(Ci-6 alkyl) is Ci_4 alkyl, such as methylene (Ci alkyl). ethylene (C 2 alkyl), or propylene (C 3 alkyl), or butylene (C 4 alkyl), more typically a Cj alkyl or C 2 alkyl, most typically a C 1 alkyl.
  • the Ci -6 alkyl of the aryl(Ci -6 alkyl), heteroaryl(Cj. 6 alkyl), heterocycly ⁇ C ⁇ alkyl) may be substituted so as to form a branched hydrocarbon.
  • R 12 , R 13 , and R 14 are an optionally substituted benzyl.
  • One of R 12 , R 13 , and R 14 may be a 4-halo-benzyl group and one may be a 4-alkoxy-benzyl group.
  • the 4-halo-benzyl group is typically a 4-fluoro- benzyl.
  • the 4-alkoxy-benzyl group is typically a C 2 - 5 alkoxybenzyl or optionally fluorinated 4-methoxy-benzyl group such as a fluoromethoxy-benzyl, difluoromethoxy- benzyl, trifluoromethoxy-benzyl group, and 2,2,2-trifluorethoxy-benzyl.
  • the compounds of the invention may be selected from the group consisting of (i) l-oxa-4,9-diaza-spiro[5.5]undecan-3-one; (ii) l-oxa-3,8-diaza- spiro[4.5]decan-2-one; (iii) l,3,8-triaza-spiro[4.5]decan-2-one; (iv) 1,2,9-triaza- spiro[5.5]undecan-3-one; (v) l,2,8-triaza-spiro[4.5]decan-3-one; (vi) 1,2, 8-triaza- spiro[4.5]decan-3-one (vii) l,2,4,8-tetraaza-spiro[4.5]decan-3-one; (viii)2,4,9-triaza- spiro[5.5]undecan-3-one; (ix) 2,8-diaza-spiro[4.5]decan-3-one (x)
  • Suitable embodiments of the compounds of Formula III may be selected from the group consisting of:
  • the compounds of Formula I, II, and III exhibit activity at monoamine receptors, specifically serotonin receptors. Certain compounds share the common property of acting as inverse agonists at the 5-HT2A receptor. Thus, experiments performed on cells transiently expressing the human phenotype of the receptor have shown that the compounds of general Formula I, II, and III attenuate the signaling of such receptors in the absence of additional ligands acting upon the receptor. The compounds have thus been found to possess intrinsic activity at this receptor and are able to attenuate the basal, non- agonist-stimulated, constitutive signaling responses that the 5-HT2A receptor displays.
  • some embodiments include use of compounds of Formula I, II, III and the salts and stereoisomers thereof, including compounds that show a relatively high degree of selectivity towards the 5-HT2A subtype of serotonin receptors relative to other subtypes of the serotonin (5-HT) family of receptors as well as to other receptors, most particularly the monoaminergic G-protein coupled receptors, such as dopamine receptors.
  • these compounds act as inverse agonists and/or antagonists at the 5-HT2A subtype of serotonin receptors.
  • the compounds of Formulae I and II may in general be prepared by routes such as those summarized below, or by modification of these methods. Ways of modifying the methodology include, among others, temperature, solvent, reagents etc, and will be obvious to those skilled in the art.
  • compounds of the Formula C may be synthesized from the corresponding ketone A by reductive animation utilizing any primary amine.
  • the reaction is conveniently carried out by stirring the reactants in an inert solvent such as methanol or ethanol containing acetic acid.
  • an inert solvent such as methanol or ethanol containing acetic acid.
  • reducing agent NaBH 4 , NaCNBHb, BH 3 -pyridine or any related reagent may be used including solid-supported reagents.
  • the reaction is typically carried out at room temperature.
  • the ketone A as exemplified by the piperidone, may be chosen from a list of compounds corresponding to the Z and Zi- groups listed in Formulae I and II.
  • the ketones can either be obtained commercially or synthesized by methodology disclosed in Lowe et al. J. Med.
  • the protecting group P includes groups such as those described in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Chemistry, 3. Ed. John Wiley & Sons, 1999, and they should be chosen in such a way, that they are stable to the reaction conditions applied and readily removed at a convenient stage using methodology known from the art.
  • Typical protecting groups are N-Boc, N-Cbz, N-Bn.
  • the amine C can be synthesized from the primary amine B by reductive amination with any aldehyde.
  • the reaction is conveniently carried out by stirring the reactants in an inert solvent such as methanol or ethanol containing acetic acid.
  • an inert solvent such as methanol or ethanol containing acetic acid.
  • reducing agent NaBH 4 , NaCNBHa, BH 3 -pyridine or any related reagent may be used including solid-supported reagents.
  • the reaction is typically carried out at room temperature.
  • the primary amine B as exemplified by the 4-aminopiperidine, may be chosen from a list of compounds corresponding to the Z and Zi-groups listed in Formulae I and II.
  • the amines can either be obtained commercially or synthesized from the corresponding ketones.
  • the protecting group P may be chosen as stated above.
  • the amine C can be synthesized from the primary amine B by alkylation with any alkylating agent (R-L 1 ).
  • the leaving group Li is suitably a halogen atom, e.g., bromine or iodine, or a sulfonate, e.g. tosylate or mesylate, or another leaving group favoring the reaction.
  • the reaction is conveniently carried out by stirring the reagents under basic conditions in an inert solvent, e.g., diisopr ⁇ pylethylamine in acetonitrile, or K2CO3 in ⁇ N-dimethylformamide.
  • the reaction is typically carried out at temperatures between room temperature and 8O 0 C.
  • the primary amine B as exemplified by the 4-aminopiperidine, may be chosen from a list of compounds corresponding to the Z and Zl -groups listed in Formulae I and II.
  • the amines can either be obtained commercially or synthesized from the corresponding ketones.
  • the protecting group P may be chosen as stated above.
  • R and R* are defined in agreement with Formulae I and II, and P represents a suitable protecting group, and Li represents a suitable leaving group.
  • the reaction is typically carried out by stirring the reactants, using an excess of isocyanate or isothiocyanate in an inert solvent, e.g., dichloromethane at a temperature between 0 0 C and room temperature and under dry conditions.
  • the reaction is typically carried out using an excess of the acylating agent and a suitable base, e.g., triethylamine or diisopropylethylamine in an inert solvent, e.g., dichloromethane, at a temperature between O 0 C and room temperature and under dry conditions.
  • a suitable base e.g., triethylamine or diisopropylethylamine in an inert solvent, e.g., dichloromethane
  • the amine C may be acylated using a carboxylic acid (Q 2 COOH) and a suitable coupling reagent e.g. PyBroP, DCC or EDCI.
  • the reaction is typically carried out using an excess of the acylating agent and the coupling reagent in an inert solvent, e.g., dichloromethane at a temperature between O 0 C and room temperature and under dry conditions.
  • an inert solvent e.g., dichloromethane
  • the compounds of the general structure (E) may be converted into the corresponding thioamides using methodology disclosed in Varma et al., Org. Lett. 1: 697-700 (1999); Cherkasov et al. Tetrahedron 41:2567 (1985); or Scheibye et al, Bull. Soc. Chim. BeIg. 87:229 (1978).
  • R, Qi, Q 2 , W and Wi are defined in agreement with Formulae I and II, P represents a suitable protecting group, and X represents a halide.
  • the substituent T on the ring nitrogen in compounds F or G can be introduced by a two step procedure.
  • the protecting group on the urea D or the amide E is removed using well-known methods.
  • the N-Boc group is removed by treating the protected compound with 4 M HCl in dioxane or trifluoroacetic acid in dichloromethane.
  • the reaction is conveniently carried out by stirring the reactants in an inert solvent such as methanol or ethanol.
  • solid-supported borohydride NaBH 4 , NaCNBH 3 , BH 3 -pyridine, H2/Pd-C or any related reagent may be used, including solid-supported reagents.
  • the reaction is typically carried out at room temperature.”
  • the compounds F and G can be synthesized from the secondary amine obtained from D or E as described above by alkylation with any alkylating agent (T- Lj).
  • the leaving group Li is suitably a halogen atom, e.g., bromine or iodine, or a sulfonate, e.g., tosylate or mesylate, or another leaving group favoring the reaction.
  • the reaction is conveniently carried out by stirring the reagents under basic conditions in an inert solvent, for example diisopropylethylamine in acetonitrile, or K. 2 CO 3 in NjN-dimethylforrnamide.
  • the reaction is typically carried out at temperatures between room temperature and 8O 0 C.
  • the T-group can be introduced in the first step of the synthetic sequence leading to the compounds in accordance with the present invention by JV-alkylation of compound H with any alkylating agent (T- Li).
  • the leaving group L 1 is suitably a halogen atom, e.g., bromine or iodine, or a sulfonate, e.g., tosylate or mesylate, or another leaving group favoring the reaction.
  • the reaction is conveniently carried out by stirring the reagent under basic conditions in an inert solvent, e.g., diisopropylethylamine in acetonitrile, or K 2 C ⁇ 3 in N,N-dimethylformamide.
  • the reaction is typically carried out at temperatures between room temperature and 8O 0 C.
  • the reaction is conveniently carried out by stirring the reactants in an inert solvent such as methanol or ethanol.
  • an inert solvent such as methanol or ethanol.
  • solid-supported borohydride, NaBH 4 , NaCNBH3, BH 3 -pyridine, H 2 ZPd-C or any related reagent may be used, including solid-supported reagents.
  • the reaction is typically carried out at room temperature, but less reactive carbonyl compounds may require higher temperatures and/or the preformation of the corresponding imine under water removal before addition of the reducing agent. Removal of the protecting group gives the desired compound J.
  • the secondary amine H and H 1 as exemplified by 4-piperidone and its protected derivative, may be chosen from a list of compounds corresponding to the Z and Zl -groups listed in Formulae I and II.
  • the amines can either be obtained commercially or synthesized from methodology disclosed in Lowe et al., J. Med. Chem. 37:2831-40 (1994); and Carroll et al., J. Med. Chem. 35:2184-91 (1992).
  • compounds of the general structure J may be synthesized starting from K using the method disclosed in: Kuehne et al., J. Org. Chem. 56:2701 (1991); and Kuehne et al., J. Org. Chem. (1991), 56:513.
  • R, Q 1 , Q 2 , W, and T are defined in agreement with Formulae I and II, and Li is a suitable leaving group.
  • Heterocyclylalkyl alkylating agents such as T-Li may be commercially available or are typically obtained by alkylation of a heterocycle with a bifunctional alkyl- linker, as shown below.
  • the leaving groups Li and L 2 are suitably a halogen atom, e.g., chlorine, bromine or iodine, or a sulfonate, e.g., tosylate or mesylate, or another leaving group favoring the reaction.
  • the reaction is conveniently carried out by stirring the reagent under basic conditions in an inert solvent, e.g., diisopropylethylamine in acetonitrile, or K 2 CO 3 in NvN-dimethylformamide.
  • the reaction is typically carried out at temperatures between room temperature and 8O 0 C.
  • the alkylating agent hence obtained can be either reacted in situ in the next step with the secondary amine (i.e. deprotected D/E, or H) or isolated from the reaction mixture before its further use.
  • Heterocyclylalkyl alcohols such as T + -CH 2 OH or T-OH may also be converted into suitable alkylating agents T-Li by transforming the hydroxyl into a leaving group, e.g. by tosylation, mesylation or halogenation.
  • a ⁇ 4 may either be obtained commercially or synthesized from methodology disclosed in the literature.
  • the introduction of substituents on Ari, Ar 2> A r3 and/or Ar 4 may be performed from a suitable precursor at any appropriate stage of the preparation of the compounds.
  • compounds containing an alkoxy substituents may be typically prepared by Williamson ether synthesis from the corresponding hydroxyaryl derivatives.
  • Structures bearing an amine substituent on Ari, Ar 2, A r3 and/or Ar 4 may be obtained from a suitable halo- or pseudohalo precursor (e.g. Br, I-, Cl-, triflate-, nonaflate-, tosylate-substituted aryl derivatives) by metal-catalyzed amination chemistries, such as Pd- or Ni- (Hartwig, Angew. Chem. Int. Ed., 1998, 37, 2046-2067; Yang & Buchwald, J.
  • a suitable halo- or pseudohalo precursor e.g. Br, I-, Cl-, triflate-, nonaflate-, tosylate-substituted aryl derivatives
  • metal-catalyzed amination chemistries such as Pd- or Ni- (Hartwig, An
  • the structures bearing an amide substituent on Arj, Ar 2 , A r 3 and/or Ar 4 may be obtained from a suitable halo- or pseudohalo precursor either by Pd catalyzed (Yin & Buchwald, J. Am. Chem. Soc, 2002, 124, 6043-6048) or by Cu catalyzed (Buchwald et al, J. Am. Chem. Soc, 2002, 124, 7421-7428) amidation chemistries.
  • these compounds may also be obtained from the corresponding aniline precursors either by acylation (Wolf, Liebigs Ann. Chem., 1952, 576, 35; Yasukara et al, J. Chem. Soc.
  • Compounds that carry an alkylsulfanyl substituent on Arj, Ar ⁇ A r 3 and/or Ar 4 be obtained from a suitable halo- or pseudohalo precursor by Pd catalyzed (Li, J. Org. Chem., 2002, 67, 3643-3650), or Cu catalyzed (Kwong & Buchwald, Org. Lett., 2002, 4, 3517-3520) thioetherification chemistry.
  • these compounds may be prepared by alkylation of corresponding benzenethiol precursors (Vogel, J. Chem. Soc, 1948, 1809; Landini & Rocca, Synthesis, 1974, 565-566; Bun-Hoi et al, J.
  • alkylarylsulfanyls may be obtained by irradiation of benzenethiols and alkenes (Screttas & Micha-Screttas, J. Org. Chem., 1978, 43, 1064- 1071).
  • Compounds that bear an acyl group on An, Ar 2, A ⁇ and/or Ar 4 may be prepared from corresponding aryl iodides by Pd catalyzed (Cacchi et al, Org. Lett, 2003, 5, 289-293) acylation chemistry.
  • benzenes may be obtained from the corresponding benzenes by Friedel-Crafts chemistry (Read, J. Am. Chem. Soc, 1922, 44, 1746-1755), or by addition of aryl-Grignard reagents to nitriles (Whitmore et al, J. Am. Chem. Soc, 1947, 69, 235-237) or to acyl chlorides (Whitmore & Lester, J. Am. Chem. Soc, 1942, 64, 1247), or by either Pd-catalyzed (Goo ⁇ en & Ghosh, Angew. Chem. Int. Ed. Engl., 2001, 40, 3458-3460) or Rh-catalyzed acylation of arylboronic acids.
  • Friedel-Crafts chemistry Read, J. Am. Chem. Soc, 1922, 44, 1746-1755
  • aryl-Grignard reagents to nitriles
  • the compounds of Formula III may in general be prepared by routes such as those summarized below, or by modification of these methods. Cyclization of the appropriate intermediates may be generally achieved with phosgene or its analogues such as CDI, with chloroacetylchloride or equivalents thereof or by treatment with carbondisulf ⁇ de and subsequent oxidation.
  • 4,5-disubstituted or 3,4,5-trisubstituted spirocycles may be prepared from an appropriate ⁇ -ketoester by reaction with a halide to introduce the 5-substituent, reductive amination (with a primary amine for the 3- substituent), treatment with allyl magnesium bromide, cyclisation as described above, oxidative cleavage of the double bonds (e.g. by ozonolysis) and formation of the piperidine ring by reductive amination.
  • the nitrile may be transformed into an ester, which is reacted with allyl magnesium bromide, followed by oxidative cleavage of the two olefins, formation of the piperidine by reductive amination.
  • Alkylation, deprotection of the hydrazide and cyclisation gives the desired spirocycles.
  • these steps may be inversed and additional protection steps of functional groups may be required.
  • the compounds may be obtained from an appropriate ⁇ - ketoester by reaction with a bis(2-chloroethyl) amine derivative to form the piperidine ring. Reductive amination, saponification and Curtius rearrangement lead to the cyclic urea derivative.
  • a nitroaldol reaction may be used to obtain the desired intermediate 1,2-aminoalcohol after reduction of the nitro-group, followed by cyclisation.
  • the compounds may be prepared by epoxidation of an appropriate olefin, obtained by Wittig or Homer- Wads worth-Emmons reactions from suitably protected 4-piperidone.
  • an enantioselective modification may include an asymmetric epoxidation method of the olefin as described in the literature, e.g. Jacobsen or others.
  • Sharpless asymmetric dihydroxylation method may be used followed by epoxide ring formation.
  • suitably protected 4-methoxycarbonyl- methylenepiperidine may be reduced to the allyl alcohol which is subjected to Sharpless asymmetric epoxidation according to literature procedure. Epoxide opening with a metallorganic reagent and oxidation of the resulting primary alcohol leads to the ⁇ - hydroxy carboxylic acid, which is converted into the desired spirocyclic enantiomer as described in herein.
  • suitably protected 4-methoxycarbonyl- methylenepiperidine may be converted to an enantiomerically pure epoxide by Jacobsen epoxidation, followed by ring opening with ammonia or an appropriate amine, reaction with a metallorganic reagent with the ester group, reduction, cyclisation, alkylation if required and introduction of the piperidine substituent.
  • the stereocenter may be introduced by using an appropriate ⁇ -amino acid ester as the chiral template.
  • compositions comprising a compound as described above, and a physiologically acceptable carrier, diluent, or excipient, or a combination thereof.
  • composition refers to a mixture of a compound disclosed herein with other chemical components, such as diluents or carriers.
  • the pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to, oral, intramuscular, intraocular, intranasal, intravenous, injection, aerosol, parenteral, and topical administration.
  • compositions can also be obtained by reacting compounds with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • Pharmaceutical compositions will generally be tailored to the specific intended route of administration.
  • physiologically acceptable defines a carrier or diluent that does not abrogate the biological activity and properties of the compound.
  • compositions described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or suitable carriers or excipient(s).
  • suitable carriers or excipient(s) include butylene glycol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, s thereof.
  • Techniques for formulation and administration of the compounds of the instant application may be found in "Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA 3 18th edition, 1990, which is hereby incorporated by reference in its entirety.
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal, intraocular injections or as an aerosol inhalant.
  • compositions disclosed herein may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes.
  • compositions for use in accordance with the present disclosure thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations, which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., as disclosed in Remington's Pharmaceutical Sciences, cited above.
  • the agents disclosed herein may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds disclosed herein to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained by mixing one or more solid excipient with pharmaceutical combination disclosed herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally, include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the present disclosure are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents, which increases the solubility of the compounds to allow for the preparation of highly, concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • a pharmaceutical carrier for the hydrophobic compounds disclosed herein is a co-solvent system comprising benzyl alcohol, a nonpolar surfactant, a water- miscible organic polymer, and an aqueous phase.
  • a common co-solvent system used is the VPD co-solvent system, which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80TM, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • VPD co-solvent system which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80TM, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics.
  • co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of Polysorbate 80TM; the fraction size of polyethylene glycol may be varied; and other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone.
  • other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those skilled in the art.
  • Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • additional strategies for protein stabilization may be employed.
  • salts may be provided as salts with pharmaceutically compatible counterions.
  • Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free acids or base forms.
  • compositions suitable for use in the methods disclosed herein include compositions where the active ingredients are contained in an amount effective to achieve its intended purpose. More specifically, a “therapeutically effective amount” means an amount of compound effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the exact formulation, route of administration and dosage for the pharmaceutical compositions disclosed herein can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl et al. 1975, in "The Pharmacological Basis of Therapeutics", Chapter 1, which is hereby incorporated by reference in its entirety).
  • the dose range of the composition administered to the patient can be from about 0.5 to 1000 mg/kg of the patient's body weight, or 1 to 500 mg/kg, or 10 to 500 mg/kg, or 50 to 100 mg/kg of the patient's body weight.
  • the dosage may be a single one or a series of two or more given in the course of one or more days, as is needed by the patient. Where no human dosage is established, a suitable human dosage can be inferred from ED 5 0 or ID 5 0 values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals.
  • the daily dosage regimen for an adult human patient may be, for example, an oral dose of between 0.1 mg and 500 mg of each ingredient, preferably between 1 mg and 250 mg, e.g. 5 to 200 mg or an intravenous, subcutaneous, or intramuscular dose of each ingredient between 0.01 mg and 100 mg, preferably between 0.1 mg and 60 mg, e.g. 1 to 40 mg of each ingredient of the pharmaceutical compositions disclosed herein or a pharmaceutically acceptable salt thereof calculated as the free base, the composition being administered 1 to 4 times per day.
  • compositions disclosed herein may be administered by continuous intravenous infusion, preferably at a dose of each ingredient up to 400 mg per day.
  • the total daily dosage by oral administration of each ingredient will typically be in the range 1 to 2000 mg and the total daily dosage by parenteral administration will typically be in the range 0.1 to 400 mg.
  • the compounds will be administered for a period of continuous therapy, for example for a week or more, or for months or years.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety, which are sufficient to maintain the modulating effects, or minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using MEC value.
  • Compositions should be administered using a regimen, which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • compositions may, if desired, be presented in a pack or dispenser device, which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may for example comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert.
  • Compositions comprising a compound disclosed herein formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • N-((4-Methylphenyl)methyl)-N-( 1 -(phenylmethyl)piperidin-4-yl)-N'- phenylmeth ylcarbamide was prepared as described in example 9 above. A sample was precipitated as the oxalate and recrystallized from ethyl acetate to give the title compound.
  • N-((4-Methylphenyl)methyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyphenylacetamide was dissolved in ether (2 ml) and HCl (3 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2 h, water (5 ml) was added, and the mixture was extracted with HCl (0.1 N, 3 X 30 ml). The combined aqueous layers were treated with NaOH (0.2 N) until basic (pH >8). The aqueous layer was extracted with diethyl ether (2 X 20 ml).
  • N-(3-Phenyl ⁇ ropyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4-methoxyph enylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na 2 SO-O, filtered to give a clear solution.
  • N-(2-Phenylethyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4-methoxyphe nylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na 2 SO 4 ), filtered to give a clear solution.
  • N-((2-Methoxyphenyl)methyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na 2 SO 4 ), filtered to give a clear solution.
  • N-((2-Chlorophenyl)methyl)-N-(l-(tert- butyloxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na 2 SO 4 ), filtered to give a clear solution.
  • N-((3 ,4-Di-methoxyphenyl)methyl)-N-( 1 -(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na 2 SO 4 ), filtered to give a clear solution.
  • N-((4-Fluorophenyl)methyl)-N-(l-(tert- butyloxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichioromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na 2 SO 4 ), filtered to give a clear solution.
  • Example 26 N-ff2.4-Di-chlorophenvDmethyl)-N-Cpiperidin-4-vD-4- methoxyphenylacetamide f26HCH80-8)
  • N-((2,4-Di-chlorophenyl)methyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml. 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15. ml), dried (Na 2 SO 4 ), filtered to give a clear solution.
  • N-((3-Methylphenyl)methyl)-N-(l-(tert- butyloxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na 2 SO 4 ), filtered to give a clear solution.
  • N-((3-Bromophenyl)methyl)-N-(l-(tert- butyloxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na 2 SO 4 ), filtered to give a clear solution.
  • Example 34 N-((4-Methylphenyl>methyl)-N- ⁇ -piperidin-4-vn-f 4- chlorophenox y”) acetamide f26HCH78-5') [0232] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (1.75 g, 8.8 mmol) and 4-methylbenzylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (1 M, 6.7 ml) followed by NaCNBHa in methanol (0.3 M, 30 ml). The resulting solution was stirred at room temperature.
  • the mixture was sequentially extracted with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and H 2 O (10 ml), dried (Na 2 SO 4 ), filtered and concentrated.
  • the crude material was dissolved in diethyl ether (2 ml) and HCl (4 M in dioxane, 1 ml). The reaction mixture was stirred at room temperature. After 2 h, NaOH (6 N, 1 ml) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 10 ml), dried (Na2SO4), and filtered to give a clear solution.
  • N-((4-Methylphenyl)methyl)amino-l-phenylmethylpyrrolid ⁇ ne 35 mg, 0.125 mmol was dissolved in dichloromethane (1.5 ml), and benzylisocyanate (0.09 ml, 0.3 mmol) was added. The reaction mixture was stirred at room temperature. After 48 h, the crude reaction mixture was added on to a column carrying strongly acidic cation exchange resin, which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound.
  • R-SAT Receptor Selection and Amplification Technology
  • All receptor and G-protein constructs used were in the pSI mammalian expression vector (Promega Inc) as described in U.S. Pat. No. 5,707,798.
  • the 5HT2A receptor gene was amplified by nested PCR from brain cDNA using the oligodeoxynucleotides based on the published sequence (see Saltzman et. al. Biochem. Biophys. Res. Comm. 181 :1469-78 (1991)). Large-scale transfections, cells were transfected for 12-16 hours, then trypsinized and frozen in DMSO. Frozen cells were later thawed, plated at 10,000-40,000 cells per well of a 96 well plate that contained drug.
  • the graph shown in Figure 1 represents the data obtained from a dose response analysis of 26HCH17 and ritanserin as 5-HT2A receptor inverse agonists. Briefly, the 5-HT2A receptor, and the alpha subunit of the guanine nucleotide binding protein Gq were transiently transfected into NIH3T3 cells and assayed using the functional receptor assay, Receptor Selection and Amplification Technology (R-SAT) essentially as disclosed in U.S. Pat. No. 5,707,798. Each compound was screened at seven serially diluted concentrations in triplicate. Data were analyzed using least squares fit analysis with GraphPad Prism (San Diego, Calif.), and are reported normalized to percent response.
  • R-SAT Receptor Selection and Amplification Technology
  • 26HCH16D is a highly selective 5-HT2A inverse agonist.
  • Method A Agilent HPl 100 HPLC/MSD.
  • Reaction-Step 1 4-(4-Trifluoromethylbenzylamino)-l-methyl ⁇ iperidin (50ELH2).
  • Reaction-Step 2 2-(4-Fluorophenyl)-N-(4-trifluoromethylbenzyl)-N- (l-methylpiperidin-4-yl)a cetamide (50ELH14c).
  • Reaction-Step 2 2-(4-Trifluoromethylphenyl)-N-(4- trifluoromethylbenzyl)-N-(l-methylpiperid in-4-yl)acetamide (50ELH14a).
  • Reaction-Step 1 4-(4-Fluorobenzylamino)-l-methylpiperidine (50ELH4).
  • Reaction-Step 2 2-(Phenyl)-N-(4-fluorobenzyl)-N-(l-methylpiperidin- 4-yl)acetamide (50ELH10).
  • Reaction-Step 2 2-(4-Trifluoromethlphenyl)-N-(4-fluorobenzyl)-N-(l- methylpiperidin-4-yl)ac etamide (50ELH1 22).

Abstract

Inverse agonists and antagonists of serotonin receptors are disclosed for use in treating sleep disorders such as insomnia, and specifically sleep maintenance insomnia. The compound increase slow wave sleep, decrease the number of awakenings after sleep onset, and decrease the time awake after sleep onset.

Description

USE OF 4-AMINO-PIPERIDINES FOR TREATING SLEEP DISORDERS
RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Patent Application No. 60/793,741, entitled "USE OF 4-AMINO-PIPERIDINES FOR TREATING SLEEP DISORDERS," filed April 19, 2006, which is hereby incorporated by reference in its entirety, including any drawings.
BACKGROUND OF THE INVENTION Field of the Invention
[0002] The present invention relates to treating sleep disorders using compounds that are selective towards monoamine receptors such as serotonin receptors. Compounds that are useful for treating sleep disorders include 4-amino-piperidines.
Description of the Related Art
[0003] One common sleep disorder is insomnia. In some patients, the insomnia is sleep maintenance insomnia. Sleep maintenance insomnia (SMI) is the inability to stay asleep or to resume sleep after waking and is a major unmet medical need. Deep, or slow wave, sleep decreases with age, which leads to superficial sleep and difficulty staying asleep. There is also an increased incidence of SMI in medical, neurological and psychiatric conditions. Patients with SMI complain of frequent awakenings and difficulty staying asleep after falling asleep. Patients with these symptoms also frequently report impairments of daytime functioning. Most available sleep agents are sedatives that are ineffective in treating the symptoms of SMI. Thus, there is a need for new drugs to treat insomnia, and specifically, sleep maintenance insomnia.
SUMMARY OF THE INVENTION
[0004] One embodiment described herein includes a method of increasing slow- wave sleep comprising administering N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide to a subject in need of increased slow wave sleep in an amount sufficient to increase slow wave sleep. One embodiment further comprises informing the subject that N-(I -methylpiperidin-4-yl)- N-(4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide increases slow wave sleep. In one embodiment, the informing comprises providing printed matter that advises that N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide increases slow wave sleep. In one embodiment, the printed matter is a label.
[0005] Another embodiment disclosed herein includes a method of treating insomnia comprising administering N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide to a subject suffering from insomnia in an amount sufficient to ameliorate insomnia. One embodiment further comprises informing the subject that N-(l-methylpiperidiή-4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide ameliorates insomnia. In one embodiment, the informing comprises providing printed matter that N- (l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide ameliorates insomnia. In one embodiment, the printed matter is a label.
[0006] Another embodiment disclosed herein includes a method for decreasing the number of awakenings after sleep onset comprising administering N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylproρyloxy)phenylmethyl) carbamide to a subject in need of a decreased number of awakenings after sleep onset in an amount sufficient to decrease the number of awakenings after sleep onset.
[0007] Another embodiment disclosed herein includes a method for decreasing the time awake after sleep onset comprising administering N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide to a subject in need of decreased time awake after sleep onset in an amount sufficient to decrease time awake after sleep onset.
[0008] Another embodiment disclosed herein includes a method of manufacturing a pharmaceutical composition including obtaining a first dosage form comprising a first amount of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4- (2-methylpropyloxy)phenylmethyl) carbamide, obtaining a second dosage form comprising a second amount of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'- (4-(2-methylpropyloxy)phenylmethyl) carbamide, and packaging together the first dosage form and the second dosage form. In one embodiment, the first dosage form and the second dosage form each comprise an oral dosage form. In one embodiment, the first dosage form comprises a higher dose of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide than the second dosage form.
[0009] In one embodiment of any of the above methods, the amount of N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide is in the range of about 0.5 rag to about 50 mg. In one embodiment, the amount is in the range of about 1 mg to about 40 mg. In one embodiment, the amount is in the range of about 2.5 mg to about 30 mg. In one embodiment, the amount is in the range of about 5 mg to about 20 mg. In one embodiment, the administration does not affect a sleep parameter selected from the group consisting of sleep period time, total sleep time, sleep onset latency, number of stage shifts, total time awake, early morning wake, sleep efficiency index, microarousal index, and a REM sleep parameter. In one embodiment, the REM sleep parameter is selected from the group consisting of REM sleep duration, proportion of REM sleep, REM sleep latency, REM activity and REM density. In one embodiment, the amount results in a steady state blood plasma concentration of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the range of about 2 ng/mL to about 60 ng/mL. In one embodiment, the amount results in a steady state blood plasma concentration in the range of about 4 ng/mL to about 50 ng/mL. In one embodiment, the amount results in a steady state blood plasma concentration of in the range of about 6 ng/mL to about 40 ng/mL. In one embodiment, the amount results in a steady state blood plasma concentration in the range of about 8.5 ng/mL to about 35 ng/mL. In one embodiment, the subject is administered N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide once every day. In one embodiment, the subject is administered a first dosage form at least 24 hours prior to administration of a second dosage form. In one embodiment, the first dosage form comprises a higher dose than the second dosage form. In one embodiment, N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide is administered at a time other than immediately before a sleep period. In one embodiment, it is administered in the morning. [0010] Another embodiment disclosed herein includes a packaged pharmaceutical composition, comprising N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in a container and instructions for using N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide to treat insomnia.
[00111 Another embodiment disclosed herein includes a packaged pharmaceutical composition, comprising N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in a container and instructions for using N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide to increase slow wave sleep, decrease the number of awakenings after sleep onset or decrease the time awake after sleep onset.
[0012] Another embodiment disclosed herein includes a kit comprising a first dosage form of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)ρhenylmethyl) carbamide and a second dosage form of N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide. In one embodiment, the first dosage form contains a higher dose of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide than the second dosage form. In one embodiment, the kit further comprises instructions for taking the first dosage form at least 24 hours before taking the second dosage form. In one embodiment, the instructions are on a label. In one embodiment, the instructions further inform a subject that N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide increases slow wave sleep. In one embodiment, the instructions further inform a subject that N-(l-methylpiperidin-4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide ameliorates insomnia. In one embodiment, the instructions further inform a subject that N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide decreases the number of awakenings after sleep onset. In one embodiment, the instructions further inform a subject that N-(I- methylpiperidin-4-yl)-N-(4-fϊuorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide decreases the time awake after sleep onset.
BRIEF DESCRIPTION OF THE DRAWINGS [0013] FIGURE 1 is a graph depicting the change in slow wave sleep duration from baseline for placebo and various doses of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide.
[0014] FIGURE 2 is a graph depicting the correlation between slow wave sleep duration and blood plasma level of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N ' -(4-(2-methylpropyloxy)phenylmethyl) carbamide.
[0015] FIGURE 3 is a graph depicting the change in number of awakenings from baseline for placebo and various doses of N-(l-methylρiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT [0016] The present application relates to the use of compounds that are inverse agonists or antagonists of a serotonin receptor to treat a sleep disorder. In some embodiments, the compounds are inverse agonists or antagonists of a 5-HT2A receptor. In some embodiments, the compounds are selective inverse agonists or antagonists. In some embodiments, the sleep disorder is insomnia. In some embodiments, the insomnia is sleep maintenance insomnia. In some embodiments, the sleep maintenance insomnia is caused by other disorders. For example, in some embodiments, the sleep maintenance insomnia is comorbid with a psychiatric disorder including, but not limited to, schizophrenia, affective disorders including MDD, bipolar 2 depression, bipolar 1 mania, rapid cyclers, dysthymia, PTSD (e.g., nightmares or arousals), alcoholism, substance abuse, drug withdrawal, and anxiety. In some embodiments, the compounds described herein may be administered in combination with an SSRI or NASR to decrease insomnia, increase efficacy, decrease sexual side effects and akathisia, provide a faster response, and limit treatment resistance. In other embodiments, the sleep maintenance insomnia is caused by a neurological disorder, including but not limited to Parkinson's disease, multisystems atrophy, migraines, multiple sclerosis, Huntington's Chorea, "Sun-downing" in DAT and other dementia's, and epilepsy. Other disorders that may cause sleep maintenance insomnia that can be treated using the compounds described herein include, but are not limited to, rheumatoid and osteo-arthritis and other chronic disorders with pain, fibro-myalgia, and female menopause.
[0017] In some embodiments, the compounds described herein are administered to a patient to increase slow wave sleep in the patient for any purpose. In some embodiments, the compounds decrease the number of awakenings after sleep onset and the time awake after sleep onset.
[0018] In some embodiments, the compounds described herein are administered to a patient to treat other sleep related disorders including periodic limb movement syndrome or obstructive sleep apnea.
[0019] In some embodiments, the compounds achieve the desired effect on slow wave sleep, number of awakenings after sleep onset, and the time awake after sleep onset without affecting other sleep parameters. In some embodiments, sleep parameters unaffected include sleep period time, total sleep time, sleep onset latency, number of sleep stage shifts, total time awake, early morning wake, sleep efficiency index, microarousal index, and REM sleep parameters (e.g., REM sleep duration, proportion of REM sleep, REM sleep latency, REM activity, and REM density).
[0020] In some embodiments, the compounds are administered at a dose between about 0.5 mg and about 50 mg, between about 1 mg and about 40 mg, between about 2.5 mg and about 30 mg, or between about 1 mg and about 20 mg. In some embodiments, the dosage and administration regiman are sufficient to achieve a steady state blood plasma concentration of between about 2 ng/ml and about 60 ng/ml, between about 4 ng/ml and about 50 ng/ml, between about 6 ng/ml and about 40 ng/ml, or between about 8.5 ng/ml and about 35 ng/ml.
[0021] In some embodiments, the dosage is varied over the dosage regimen. For example, in one embodiment, the first administration or series of administrations have a higher dosage than subsequent administrations. In some embodiments, the compound administered has a relatively high half-life such that after an initial dosage sufficient to raise the blood plasma concentration to a desired level, subsequent dosages sufficient to maintain a desired steady state blood plasma concentration can be lower. In some embodiments, pharmaceutical compositions comprising multiple doses are packaged together.
[0022] In some embodiments, a sleep-inducing agent is administered in combination with the compounds described herein. The sleep-inducing agent may be administered to induce onset of sleep in the patient while the inverse agonist or antagonist of a serotonin receptor may be administered to maintain sleep in the patient. For example, in some embodiments, the compounds described herein may be administered to maintain slow wave sleep in the patient. Non-limiting examples of suitable sleep-inducing agents include AMBIEN®, indiplon, LUNESTA®, and melatonin.
[0023] By administration in "combination," it is meant that the two or more agents may be found in the patient's bloodstream at the same time, regardless of when or how they are actually administered. In one embodiment, the agents are administered simultaneously. In one such embodiment, administration in combination is accomplished by combining the agents in a single dosage form. In another embodiment, the agents are administered sequentially. In one embodiment the agents are administered through the same route, such as orally. In another embodiment, the agents are administered through different routes, such as one being administered orally and another being administered intravenously. In one advantageous embodiment, the pharmacokinetics of the two or more agents are substantially the same.
[0024] In some embodiments, the compounds described herein are long acting (e.g., have a long half-life), allowing them to be given relatively infrequently and at times other than immediately prior to a desired sleep period. For example, in some embodiments, the compounds have a half-life long enough so that that they don't need to be administered more often than once a day. In various embodiments, the compound may be administered once every 1, 2, 3, 4, or 5 days and still provide sleep maintenance properties during each sleep period. In various embodiments, the half-life of the compound is from about 10 hours to about 100 hours, from about 20 hours to about 60 hours, or from about 35 hours to about 55 hours. In some embodiments, the compound is not administered immediately prior to a sleep period. For example, the compound may be administered in the morning or afternoon. In one embodiment, the compound is administered in the morning. In some embodiments, the long acting properties of the compounds eliminate or reduce any withdrawal effects experienced by patients as either a co-administered sleep-inducing agent or the inverse agonist or antagonist of a serotonin receptor wears off. For example, many short acting sleep agents cause a patient to wake in the middle of their sleep period and experience a withdrawal effect. In some embodiments, patients taking a compound described herein experience no such withdrawal effect. Definitions
[0025] For the purpose of the current disclosure, the following definitions shall in their entireties be used to define technical terms, and shall also, in their entireties, be used to define the scope of the composition of matter for which protection is sought in the claims. The term "constitutive activity" is defined as the basal activity of a receptor which is independent of the presence of an agonist. Constitutive activity of a receptor may be measured using a number of different methods, including cellular (e.g., membrane) preparations (see, e.g., Barr &. Manning, J. Biol. Chem. 272:32979-87 (1997)), purified reconstituted receptors with or without the associated G-protein in phospholipid vesicles (Cerione et al., Biochemistry 23:4519-25 (1984)), and functional cellular assays (U.S. Patent 6,358,698).
[0026] The term "agonist" is defined as a compound that increases the activity of a receptor when it contacts the receptor.
[0027] The term "antagonist" is defined as a compound that competes with an agonist or inverse agonist for binding to a receptor, thereby blocking the action of an agonist or inverse agonist on the receptor. However, an antagonist (also known as a "neutral" antagonist) has no effect on constitutive receptor activity.
[0028] The term "inverse agonist" is defined as a compound that decreases the basal activity of a receptor (i.e., signaling mediated by the receptor). Such compounds are also known as negative antagonists. An inverse agonist is a ligand for a receptor that causes the receptor to adopt an inactive state relative to a basal state occurring in the absence of any ligand. Thus, while an antagonist can inhibit the activity of an agonist, an inverse agonist is a ligand that can alter the conformation of the receptor in the absence of an agonist. The concept of an inverse agonist has been explored by Bond et al. in Nature 374:272 (1995). More specifically, Bond et al. have proposed that unliganded p2- adrenoceptor exists in an equilibrium between an inactive conformation and a spontaneously active conformation. Agonists are proposed to stabilize the receptor in an active conformation. Conversely, inverse agonists are believed to stabilize an inactive receptor conformation. Thus, while an antagonist manifests its activity by virtue of inhibiting an agonist, an inverse agonist can additionally manifest its activity in the absence of an agonist by inhibiting the spontaneous conversion of an unliganded receptor to an active conformation.
[0029] The term "5-HT2A receptor" is defined as a receptor, having an activity corresponding to the activity of the human serotonin receptor subtype, which was characterized through molecular cloning and pharmacology as detailed in Saltzman et al., Biochem. Biophys. Res. Comm. 181:1469-78; and Julius et al., Proc. Natl. Acad. ScL USA 87:928-932.
[0030] The term "subject" refers to an animal, preferably a mammal, most preferably a human, who is the object of treatment, observation or experiment.
[0031] The term "selective" is defined as a property of a compound whereby an amount of the compound sufficient to effect a desired response from a particular receptor type, subtype, class or subclass causes a substantially smaller or no effect upon the activity other receptor types.
[0032] The terms "selectivity" or "selective," in relation to an inverse agonist, are understood as a property of a compound of the invention whereby an amount of compound that effectively inversely agonizes the 5 -HT2 A. receptor, and thereby decreases its activity, causes little or no inverse agonistic or antagonistic activity at other, related or unrelated, receptors. In particular, certain compounds of the invention have been found not to interact strongly with other serotonin receptors (S-HT IA, IB, ID, IE, IF, 2B, 2C5 4 A, 6, and 7) at concentrations where the signaling of the 5-HT2 A receptor is strongly or completely inhibited. Preferably, the compounds of the invention are also selective with respect to other monoamine-binding receptors, such as the dopaminergic, histaminergic, adrenergic and muscarinic receptors. Compounds that are highly selective for 5-HT2A receptors may have a beneficial effect in the treatment of psychosis, schizophrenia or similar neuropsychiatric disorders, while avoiding adverse effects associated with drugs hitherto suggested for this purpose.
[0033] The EC50 for an agonist is intended to denote the concentration of a compound needed to achieve 50% of a maximal response seen in R-SAT. For inverse agonists, EC50 is intended to denote the concentration of a compound needed to achieve 50% inhibition of an R-SAT response from basal, no compound, levels.
[0034] In the present context the term "aryl" is intended to mean a carbocyclic aromatic ring or ring system. Moreover, the term "aryl" includes fused ring systems wherein at least two aryl rings, or at least one aryl and at least one C3-8-cycloalkyl share at least one chemical bond. Some examples of "aryl" rings include optionally substituted phenyl, naphthalenyl, phenanthrenyl, anthracenyl, tetralinyl, fluorenyl, indenyl, and indanyl. The term "aryl" relates to aromatic, preferably benzenoid groups, connected via one of the ring-forming carbon atoms, and optionally carrying one or more substituents selected from heterocyclyl, heteroaryl, halo, hydroxy, amino, cyano, nitro, alkylamido, acyl, Ci-6 alkoxy, Ci-6 alkyl, Ci-6 hydroxyalkyl, Ci-6 aminoalkyl, Ci-e alkylamino, alkylsulfenyl, alkylsulfϊnyl, alkylsulfonyl, sulfamoyl, or trifluoromethyl. The aryl group may be substituted at the para and/or meta positions. Representative examples of aryl groups include, but are not limited to, phenyl, 3-halophenyl, 4-halophenyl, 3- hydroxyphenyl, 4-hydroxyphenyl, 3-aminophenyl, 4-aminophenyl, 3-methylphenyl, 4- methylphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 4-trifluoromethoxyphenyl 3- cyanophenyl, 4-cyanophenyl, dimethylphenyl, naphthyl, hydroxynaphthyl, hydroxymethylphenyl, trifluoromethylphenyl, alkoxyphenyl, 4-moφholin-4-ylphenyl, 4- pyrrolidin-1-ylphenyl, 4-pyrazolylphenyl, 4-triazolylphenyl, and 4-(2-oxopyrrolidin-l- yl)phenyl.
[0035] In the present context, the term "heteroaryl" is intended to mean a heterocyclic aromatic group where one or more carbon atoms in an aromatic ring have been replaced with one or more heteroatoms selected from the group comprising nitrogen, sulfur, phosphorous, and oxygen.
[0036] Furthermore, in the present context, the term "heteroaryl" comprises fused ring systems wherein at least one aryl ring and at least one heteroaryl ring, at least two heteroaryl rings, at least one heteroaryl ring and at least one heterocyclyl ring, or at least one heteroaryl ring and at least one C3.s-cycloalkyl ring share at least one chemical bond.
[0037] The term "heteroaryl" is understood to relate to aromatic, C3-8 cyclic groups further containing one oxygen or sulfur atom or up to four nitrogen atoms, or a combination of one oxygen or sulfur atom with up to two nitrogen atoms, and their substituted as well as benzo- and pyrido-fused derivatives, preferably connected via one of the ring-forming carbon atoms. Heteroaryl groups may carry one or more substituents, selected from halo, hydroxy, amino, cyano, nitro, alkylamido, acyl, Ci-6-alkoxy, C1-6- alkyl, Cj-6-hydroxyalkyl, Ci-6-aminoalkyl, Ci.6-alkylamino, alkylsulfenyl, alkylsulfinyl, alkylsulfonyl, sulfamoyl, or trifluoromethyl. In some embodiments, heteroaryl groups may be five- and six-membered aromatic heterocyclic systems carrying 0, 1, or 2 substituents, which may be the same as or different from one another, selected from the list above. Representative examples of heteroaryl groups include, but are not limited to, unsubstituted and mono- or di-substituted derivatives of furan, benzofuran, thiophene, benzothiophene, pyrrole, pyridine, indole, oxazole, benzoxazole, isoxazole, benzisoxazole, thiazole, benzothiazole, isothiazole, imidazole, benzimidazole, pyrazole, indazole, tetrazole, quionoline, isoquinoline, pyridazine, pyrimidine, purine and pyrazine, which are all preferred, as well as furazan, 1,2,3-oxadiazole, 1,2,3-thiadiazole, 1,2,4- thiadiazole, triazole, benzotriazole, pteridine, phenoxazole, oxadiazole, benzopyrazole, quinolizine, cinnoline, phthalazine, quinazoline, and quinoxaline. In some embodiments, the substituents are halo, hydroxy, cyano, O-Ci-6-alkyl, Ci^-alkyl, hydroxy-Ci-β-alkyl, amino-Ci.6-al.cyl.
[0038] In the present context, the term "alkyl", "d-6-alkyl" are intended to mean a linear or branched saturated hydrocarbon chain wherein the longest chain has from one to six carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec- butyl, tert-butyl, pentyl, isopentyl, neopentyl, and hexyl. An alkyl chain may be optionally substituted.
[0039] "Lower alkyl groups" are Ci-6 cyclic, straight-chained or branched aliphatic substituent groups connected via a carbon atom. Examples include methyl, ethyl, propyl, butyl, methylbutyl, cyclopropyl, cyclohexyl, iso-propyl, tert-butyl.
[0040] In the present context, the term "C2-8-alkenyl" is intended to mean a linear or branched hydrocarbon group having from two to eight carbon atoms and containing one or more double bonds. Some examples of C2-8-alkenyl groups include allyl, homo-allyl, vinyl, crotyl, butenyl, pentenyl, hexenyl, heptenyl and octenyl. Some examples of C2-8-alkenyl groups with more than one double bond include butadienyl, pentadienyl, hexadienyl, heptadienyl, heptatrienyl and octatrienyl groups as well as branched forms of these. The position of the unsaturation (the double bond) may be at any position along the carbon chain.
[0041] In the present context the term "C2-8-alkynyl" is intended to mean a linear or branched hydrocarbon group containing from two to eight carbon atoms and containing one or more triple bonds. Some examples of C2-8-alkynyl groups include ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl and octynyl groups as well as branched forms of these. The position of unsaturation (the triple bond) may be at any position along the carbon chain. More than one bond may be unsaturated such that the "C2-8-alkynyl" is a di-yne or enedi-yne as is known to the person skilled in the art.
[0042] In the present context, the term "Cs-s-cycloalkyl" is intended to cover three-, four-, five-, six-, seven-, and eight-membered rings comprising carbon atoms only. A Ca-β-cycloalkyl may optionally contain one or more unsaturated bonds situated in such a way, however, that an aromatic π-electron system does not arise. [0043] Some examples of preferred "Ca^-cycloalkyl" are the carbocycles cyclopropane, cyclobutane, cyclopentane, cyclopentene, cyclopentadiene, cyclohexane, cyclohexene, 1,3-cyclohexadiene, 1,4-cyclohexadiene, cycloheptane, cycloheptene.
[0044] "Cyclic organyl groups" are aliphatic, alicyclic groups in which carbon atoms form a ring. In preferred embodiments containing three, four, five, six or seven carbon atoms, the ring, as a substituent, is connected either directly via one of the ring atoms or via one or more appended carbon atoms. Particular examples of such groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, cyclohexylethyl groups, and the like.
[0045] "Straight-chained acyclic organyl groups" are substituent groups consisting of a linear arrangement of carbon atoms, where accordingly each carbon atom binds a maximum of two other carbon atoms, connected through single, double, or triple bonds. The straight-chained organyl groups may contain none, one, or several multiple bonds, and are, for example, commonly referred to as alkyl, alkenyl or alkynyl, or alkadienyl groups, respectively. Examples of straight-chained organyl groups include methyl, ethyl, propyl, butyl, pentyl, hexyl, propenyl, butenyl, pentadienyl, propargyl, butynyl.
[0046] "Branched acyclic organyl groups" are substituent groups consisting of a branched arrangement of carbon atoms, where accordingly one or more carbon atoms may bind more than two other carbon atoms, connected through single, double, or triple bonds. The branched organyl groups may contain none, one, or several multiple bonds. Examples of branched organyl groups include iso-propyl, iso-butyl, tert-butyl, methylbutyl, methylbutenyl, methylbutynyl.
[0047] The term "C)-6 alkoxy" and "lower alkoxy groups" are understood as C ] .6 cyclic or acyclic organyl groups connected, as substituents, via an oxygen atom. Examples of lower alkoxy groups include methoxy, ethoxy, iso-propoxy, butoxy, tert- butoxy, cyclopropyl, cyclobutyl, cyclopropylmethyl, and cyclobutylmethyl.
[0048] The term "Ci.6 alkylamino" and "lower alkylamino groups" are understood as lower alkyl groups connected, as substituents, via a nitrogen atom, which may carry one or two lower alkyl groups. Particular examples include methylamino, dimethylamino, iso-propylamino. Optionally, lower aminoalkyl groups may consist of 4-6 membered nitrogen-containing rings, such as pyrrolidino. [0049] The term
Figure imgf000014_0001
aminoalkyl" and "lower aminoalkyl groups" are understood as lower alkyl groups carrying, as a substituent, an additional amino group. Examples include aminomethyl and aminoethyl.
[0050] The term "Ci-6-hydroxyalkyl" and "lower hydroxyalkyl groups" are understood as lower alkyl groups carrying, as a substituent, an additional hydroxy group. Examples include hydroxymethyl, hydroxyethyl, 2-hydroxy-2-propyl, hydroxypentyl.
[0051] The term "heterocyclyl" is intended to mean three-, four-, five-, six-, seven-, and eight-membered rings wherein carbon atoms together with from 1 to 3 heteroatoms constitute the ring. A heterocyclyl may optionally contain one or more unsaturated bonds situated in such a way, however, that an aromatic π-electron system does not arise. The heteroatoms are independently selected from oxygen, sulfur, and nitrogen.
[0052] A heterocyclyl may further contain one or more carbonyl or thiocarbonyl functionalities, so as to make the definition include oxo-systems and thio- systems such as lactams, lactones, cyclic imides, cyclic thioimides, cyclic carbamates, and the like.
[0053] Heterocyclyl rings may optionally also be fused to aryl rings, such that the definition includes bicyclic structures. Preferred such fused heterocyclyl groups share one bond with an optionally substituted benzene ring. Examples of benzo-fused heterocyclyl groups include, but are not limited to, benzimidazolidinone, tetrahydroquinoline, and methylenedioxybenzene ring structures.
[0054] Some examples of "heterocyclyls" include, but are not limited to, tetrahydrothiopyran, 4H-pyran, tetrahydropyran, piperidine, 1,3-dioxin, 1,3-dioxane, 1,4- dioxin, 1,4-dioxane, piperazine, 1,3-oxathiane, 1 ,4-oxathiin, 1,4-oxathiane, tetrahydro- 1 ,4-thiazine, 2//-l,2-oxazine , maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, morpholine, trioxane, hexahydro-1,3,5- triazine, tetrahydrothiophene, tetrahydrofuran, pyrroline, pyrrolidine, pyrrolidone, pyrrolidione, pyrazoline, pyrazolidine, imidazoline, imidazolidine, 1,3-dioxole, 1,3- dioxolane, 1,3-dithiole, 1,3-dithiolane, isoxazoline, isoxazolidine, oxazoline, oxazolidine, oxazolidinone, thiazoline, thiazolidine, and 1 ,3-oxathiolane. Binding to the heterocycle may be at the position of a heteroatom or via a carbon atom of the heterocycle, or, for benzo-fused derivatives, via a carbon of the benzenoid ring. [0055] The term "(heterocyclyl)Ci-6-alkyl" is understood as heterocyclyl groups connected, as substituents, via a lower alkylene, each as defined herein. The heterocyclyl groups of (heterocyclyl)Ci-6-alkyl groups may be substituted or unsubstituted.
[0056J The terms "(aryl)Ci-6-alkyl" and "aralkyl" are intended to mean an aryl group connected, as a substituent, via a lower alkylene, each as defined herein. The aryl groups of (ary^Ci-ή-alkyl may be substituted or unsubstituted. Examples include benzyl, substituted benzyl, 2-phenylethyl, 3-phenylpropyl, and naphthylalkyl.
[0057] The terms "(heteroaryl)Ci.6-alkyl" and "heteroaralkyl" are understood as heteroaryl groups connected, as substituents, via a lower alkylene, each as defined herein. The heteroaryl groups of heteroaralkyl groups may be substituted or unsubstituted. Examples include 2-thienylmethyl, 3-thienylmethyl, furylmethyl, thienylethyl, pyrrolylalkyl, pyridylalkyl, isoxazolylalkyl, imidazolylalkyl, and their substituted as well as benzo-fused analogs.
[0058] The terms "(cyclo alky I)C i-β-alkyl" is intended to mean a cycloalkyl groups connected, as substituents, via a lower alkylene, each as defined herein.
[0059] When used herein, the term "O-Ci-e-alkyl" is intended to mean Ci-g- alkyloxy, or alkoxy, such as methoxy. ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, pentyloxy, isopentyloxy, neopentyloxy and hexyloxy. Further, the definition of "O-Ci.6-alkyl" is intended to cover cyclic alkoxy groups having a maximum of six carbon atoms. Illustrative non-limiting examples of cyclic alkoxy groups include cyclobutyloxy, cyclopropylmethyloxy, cyclohexyloxy, and the like.
[0060] In the present context the term "lower alkylene" means a bivalent hydrocarbon tether, containing from one to six carbon atoms. Additionally, "lower alkylene" tethers may optionally contain one or more substituents selected from Ci-βalkyl, halogen, hydroxyl, and amino. Non-limiting examples of "lower alkylene" groups are methylene, ethylene, propylene, tetramethylene, hexamethylene.
[0061] "Vinylene groups" are ethene-l,2-diyl groups (— CHCH-) having (E) or (Z) configuration.
[0062] "Acyl groups" are hydrogen or lower alkyl groups connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl. [0063] The terms "haloalkyl", "hydroxyalkyl" and "aminoalkyl" are intended to cover Cj-6-alkyl groups, defined above, carrying at least one halogen, hydroxy group, or amino group, respectively.
[0064] The term "halogen" includes fluorine, chlorine, bromine and iodine.
[0065] In the present context, i.e. in connection with the terms "Ci-6-alkyl", "aryl", "heteroaryl", "heterocyclyl", "C3.8-cycloalkyl", "(aryl)Ci-6-alkyl", "(heteroaryl)C,. 6-alkyl", "(heterocyclyOCi-e-alkyl", "(cycloalkyl)Ci.6alkyl", "O-C,,6-alkyl", "C2.8- alkenyl", and "C2-8-alkynyl", the term "optionally substituted" is intended to mean that the group in question may be substituted one or several times, such as 1 to 5 times, or 1 to 3 times, or 1 to 2 times, with one or more groups selected from Ci-e-alkyl, Ci-6-alkoxy, oxo (which may be represented in the tautomeric enol form), carboxyl, amino, hydroxy (which when present in an enol system may be represented in the tautomeric keto form), nitro, alkylsulfonyl, alkylsulfenyl, alkylsulfϊnyl,Ci-6-alkoxycarbonyl, d-β-alkylcarbonyl, forrnyl, amino, mono- and di(Ci-6-alkyl)amino; carbamoyl, mono- and di(C]-6- alkyl)aminocarbonyl, amino-Ci-e-alkyl-aminocarbonyl, mono- and di(Ci-6-alkyl)amino- Ci-6-alkyl-aminocarbonyl, Ci-6-alkylcarbonylamino, C].6-alkylhydroxyimino, cyano, guanidino, carbamido, Ci-6-alkanoyloxy, Q-e-alkylsulphonyloxy, dihalogen-Ci-β-alkyl, trihalogen-Ci-6-alkyl, heterocyclyl, heteroaryl, and halo. In general, the above substituents may be susceptible to further optional substitution.
[0066] The term "salts" is intended to mean pharmaceutically acceptable acid addition salts obtainable by treating the base form of a functional group, such as an amine, with appropriate acids such as inorganic acids, for example hydrohalic acids; typically hydrochloric, hydrobromic, hydrofluoric, or hydroiodic acid; sulfuric acid; nitric acid; phosphoric acid and the like; or organic acids, for example acetic, propionic, hydroacetic, 2-hydroxypropanoic acid, 2-oxopropanoic acid, ethandioic, propanediol, butanedioic, (Z)-2-butenedioic, (E)-butenedioic, 2-hydroxybutanedioic, 2,3-dihydroxybutanedioic, 2- hydroxy-l,2,3-propanetricarboxylic, methanesulfonic, ethanesulfonic, benzenesulfonic, 4- methylbenzenesulfonic acid, cyclohexanesulfamic, 2-hydoxybenzoic, 4-amino-2- hydroxybenzoic, and other acids known to the skilled practitioner.
[0067] The present invention includes within its scope prodrugs of the compounds of this invention. In general, such prodrugs are inactive derivatives of the compounds of this invention that are readily convertible in vivo into the required compound. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Design of Prodrugs, (ed. H. Bundgaard, Elsevier, 1985). Metabolites of these compounds include active species that are produced upon introduction of compounds of this invention into the biological milieu.
[0068] Where the compounds according to the invention have at least one chiral center, they may exist as a racemate or as enantiomers. It should be noted that all such isomers and mixtures thereof are included in the scope of the present invention. Furthermore, some of the crystalline forms for compounds of the present invention may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds of the present invention may form solvates with water (i.e., hydrates) or common organic solvents. Such solvates are also included in the scope of this invention.
[0069] Where the processes for the preparation of the compounds according to the invention give rise to mixtures of stereoisomers, such isomers may be separated by conventional techniques such as preparative chiral chromatography. The compounds may be prepared in racemic form or individual enantiomers may be prepared by stereoselective synthesis or by resolution. The compounds may be resolved into their component enantiomers by standard techniques, such as the formation of diastereomeric pairs by salt formation with an optically active acid, such as (-)-di-p-toIuoyl-d-tartaric acid and/or (+)- di-p-toluoyl-1-tartaric acid, followed by fractional crystallization and regeneration of the free base. The compounds may also be resolved using a chiral auxiliary by formation of diastereomeric derivatives such as esters, amides or ketals followed by chromatographic separation and removal of the chiral auxiliary.
Compounds
[0070] One embodiment includes the use of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide, a 5-HT2A inverse agonist which has the structure:
Figure imgf000017_0001
[0071] Other compounds that are selective serotonin inverse agonists or antagonists are described in U.S. Patent Nos. 6,756,393 and 6,815,458 entitled "AZACYCLIC COMPOUNDS", filed April 7, 2003 and March 6, 2001 respectively; U.S. Patent No. 6,911,452 entitled "SPIROAZACYCLIC COMPOUNDS AS MONOAMINE RECEPTOR MODULATORS," filed Dec. 23, 2002; U.S. Patent Application Nos. 10/802,970, filed March 16, 2004, 11/417,790, filed May 3, 2006, and 11/417,782, filed May 3, 2006, each entitled "AZACYCLIC COMPOUNDS"; U.S. Patent Application Nos. 10/601,070, filed June 20, 2003 and 11/299,566, filed Dec. 12, 2005, 11/417,866, filed May 3, 2006, and 11/418,353, filed May 3, 2006, each entitled "N- SUBSTITUTED PIPERDINE DERIVATIVES AS SEROTONIN RECEPTOR AGENTS; U.S. Patent Application Nos. 11/154,08, filed June 16, 2005, 11/418,322, filed May 3, 2006, and 11/417,439, filed May 3, 2006, each entitled "SPIROAZACYCLIC COMPOUNDS AS MONOAMINE RECEPTOR MODULATORS,"; U.S. Patent Application Nos. 10/759,561 filed Jan. 15, 2004, 11/416,527, filed May 3, 2006, and 11/416,855, filed May 3, 2006, each entitled "SELECTIVE SEROTONIN 2A/2C RECEPTOR INVERSE AGONISTS AS THERAPEUTICS FOR NEURODEGENERATIVE DISEASES"; and U.S. Patent Application Nos. 11/235,558, filed Sept. 26, 2005, 11/235,381, filed Sept. 26, 2005, 11/418,341, filed May 3, 2006 and 11/417,447, filed May 3, 2006, each entitled "SYNTHESIS OF N-(4- FLUOROBENZYL)-N-(l-METHYLPIPERIDIN-4-YL)-N'-(4-(2-
METHYLPROPYLOXY)PHENYLMETHYL) CARBAMIDE AND ITS TARTRATE SALT AND CRYSTALLINE FORMS"; U.S. Patent Application No. 60/187,289 entitled "AZACYCLIC COMPOUNDS" filed Mar. 6, 2000; U.S. Patent Application No. 60/344,750 entitled "SPIROAZCYCLIC COMPOUNDS AS MONOAMINE RECEPTOR MODULATORS" filed Dec. 28, 2001; U.S. Patent Application No. 60/391 ,269 entitled "N-[HETEROCYCLYLALKY] PIPERDINE DERIVATIVES AS SEROTONIN RECEPTOR AGENTS" filed Jun. 24, 2002; U.S. Patent Application No. 60/441,406 entitled "SELECTIVE SEROTONIN 2A/2C RECEPTOR INVERSE • AGONISTS AS THERAPEUTICS FOR NEURODEGENERATIVE DISEASES" filed Jan. 16, 2003; U.S. Patent Application No. 60/614,014 entitled "SYNTHESIS OF l-(4- FLUOROBENZYL)-3-(4-ISOBUTOXYBENZYL)-l-(l-METHYLPIPERIDIN-4- YL)UREA, ITS SALTS, AND POLYMORPHS" filed Sept. 27, 2004; the disclosures of which are hereby incorporated by reference in their entirety. [0072] One embodiment includes the use of compounds of the general Formula (I):
Figure imgf000019_0001
wherein Z is
Figure imgf000019_0002
[0073] R is a hydrogen, a cyclic or straight-chained or branched acyclic organyl group, a lower hydroxyalkyl group, a lower aminoalkyl group, or an aralkyl or heteroaralkyl group; and n can be 0, 1, or 2.
[0074] X] is methylene, vinylene, or an NH or N(lower alkyl) group; and X2 is methylene , or, when X] is methylene or vinylene, X2 is methylene or a bond; or when Xj is methylene, X2 is O, S, NH, or N(lower alkyl) or a bond.
[0075] Y] is methylene and Y2 is methylene, vinylene, ethylene, propylene, or a bond; or Yj is a bond and Y2 is vinylene; or Yj is ethylene and Y2 is O, S, NH, or N(lower alkyl).
[0076] Ari and Ar2 each independently is unsubstituted or substituted aryl or heteroaryl groups.
[0077] W is oxygen or sulfur.
[0078] In some embodiments of the compounds of Formula (I), Yi is methylene and Y2 is a bond, methylene, ethylene, or vinylene; or Yi is ethylene and Y2 is O or S; and Xj is methylene and X2 is a bond, methylene, O, or S; or Xi is NH or N(lower alkyl) and X2 is methylene.
[0079] In another embodiment, Z is
Figure imgf000020_0001
and W is oxygen;
[0080] In certain embodiments, Arj and Ar2 independently are mono- or disubstituted phenyl groups.
[0081] In other embodiments, the R of Formula (I) is a hydrogen, a lower alkyl group, a cyclic organyl group, or a substituted or unsubstituted aralkyl or heteroaralkyl group; n is 1; Yi is methylene, and Y2 is a bond, methylene, ethylene, or vinylene; Xi is methylene and X2 is a bond, or Xi is NH or N(lower alkyl) and X2 is methylene; andAri and Ar2 are phenyl groups, independently /^-substituted with groups selected from lower alkyl, lower alkoxy and halogen.
[0082] In another embodiment, compounds of Formula (Ia) are used:
Figure imgf000020_0002
wherein RN is hydrogen, lower alkyl, aralkyl, or heteroaralkyl; ArL is selected from lower alkyl, lower alkoxy and halogen; ArR is selected from lower alkyl, lower alkoxy and halogen; k is 1 or 2; and A" is a suitable anion.
[0083] It will be understood that "Formula I" as used herein refers to compounds of Formulae (I) and (Ia).
[0084] Suitable embodiments of the compounds of Formula I may be selected from the group consisting, of: iV-((4-methylphenyl)methyl)-iV"-(piperidin-4-yl)-7V- phenylmethylcarbamide;
N-((4-methylphenyl)methyl)-N-(l-(2-methylpropyl)piperidin-4-yl)-Λ^- phenylmethylcarbamide; N-(l-(cyclohexylmethyl)piperidin-4-yl)-N-((4-methylphenyl)methyl)-N'- phenylmethylcarbamide;
N-((4rmethylphenyl)methyl)-N-(piperidin-4-yl)-4- methoxyphenylacetamide;
N-(l-(3,3-dimethylbutyl)piperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide; iV-(l-(cyclohexylmethyl)piperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide;
N-((4-methylphenyl)methyl)-N-( 1 -(2-methylpropyl)piperidin-4-yl)-4- ■ methoxyphenylacetamide;
N-(3-phenylpropyl)-N-(piperidin-4-yl)-4-methoxyphenylacetamide;
N-(2-phenylethyl)-N-(piperidin-4-yl)-4-methoxyphenylacetamide;
N-((2-methoxyphenyl)methyl)-N-(piperidin-4-yl)-4- methoxyphenylacetamide ;
N-((2-chlorophenyl)methyl)-N-(piperidin-4-yl)-4- methoxyphenylacetamide;
N-((3,4-di-methoxyphenyl)methyl)-N-(piperidin-4-yl)-4- methoxyphenylacetamide;
N-((4-fluorophenyl)methyl)-N-(piperidin-4-yl)-4- methoxyphenylacetamide;
N-((2,4-di-chlorophenyl)methyl)-N-(piperidin-4-yl)-4- methoxyphenylacetamide;
N-((3-methylphenyl)methyl)-N-(piperidin-4-yl)-4- methoxyphenylacetamide;
N-((3-bromophenyl)methyl)-N-(piperidin-4-yl)-4- methoxyphenylacetamide;
N-((4-methylphenyl)methyl)-N-(l-piperidin-4-yl)-phenylacetamide;
N-((4-methylphenyl)methyl)-N-(l-piperidin-4-yl)-3-phenylpropionamide;
N-((4-methylphenyl)methyl)-N-( 1 -piperidin-4-yl)-(phenylthio)acetamide;
N-((4-methylphenyl)methyl)-N-(l-ρiperidin-4-yl)-phenoxyacetamide;
N-((4-methylphenyl)methyl)-N-( 1 -piperidin-4-yl)-(4- chlorophenoxy)acetamide; N-((4-methylphenyl)methyl)-N-(l-piperidin-4-yl)-3- methoxyphenylacetamide;
N-((4-methylphenyl)methyl)-N-(l-piperidin-4-yl)-4- fluorophenylacetamide;
N-((4-methylphenyl)methyl)-N-(l -piperidin-4-yl)-2,5-di- methoxyphenylacetarnide;
N-((4-methylphenyl)methyl)-N-( 1 -piperidin-4-yl)-4- chlorophenylacetamide;
2-(4-methoxyρhenyl)--\''-(4-niethylben2yl)-iV-(piperidin-4-yl) acetamide;
2-(4-methoxyphenyl)-JV-(4-methylbenzyl)-JV-(l-methylpiperidin-4-yl) acetamide;
2-(4-methoxyphenyl)-Λ'-(4-niethylbenzyl)-iV-( 1 -ethylpiperidin-4-yl) acetamide;
2-(4-methoxyphenyl)-iV-(4-chlorbenzyl)-iV-( 1 -ethylpiperidin-4-yl) acetamide.
2-(4-methoxyphenyl)-Λr-(4-methylbenzyl)-iV:-(l-isopropylpiperidin-4-yl) acetamide;
2-(4-methoxyphenyl)-7V-(4-chlorobenzyl)-N-(piperidin-4-yl) acetamide;
2-(4-methoxyphenyl)-N-(4-chlorbenzyl)-iV-(l-cyclopentylpiperidin-4-yl) acetamide;
2-(4-methoxyphenyl)-N-(4-chlorbenzyl)-N-(l-isopropylpiperidin-4-yl) acetamide;
2-(phenyl)-N-(4-trifluoromethylbenzyl)-N-(l-methylpiperidin-4-yl) acetamide;
2-(4-fluorophenyl)-N-(4-trifluoromethylbenzyl)-N-(l-methylpiperidin-4-yl) acetamide;
2-(4-Methoxyphenyl)-N-(4-trifluoromethylbenzyl)-iV-(l-methylpiperidin- 4-yl) acetamide;
2-(4-Trifluoromethylphenyl)-iV-(4-trifluoromethylben2yl)-iV-(l- methylpiperidin-4-yl) acetamide;
2-(4-Fluorophenyl)-iV-(4-fluorobenzyl)-iV-( 1 -methylpiperidin-4-y 1) acetamide; 2-(4-Methoxyphenyl)-iV-(4-fluorobenzyl)-iV-(l-methylpiperidin-4-yl) acetamϊde;
2-(phenyl)-iV-(4-fluorobenzyl)-N-(l-methylpiperidin-4-yl) acetamide;
2-(4-Trifluoromethylphenyl)-iV-(4-fluorobenzyl)-N-(l-methylpiperidin-4- yl) acetamide;
2-(4-trifluoromethylphenyl)-N-[4-(methoxycarbonyl)benzyl]-N-( 1 - methylpiperidin-4-yl) acetamide;
2-(4-Methoxyphenyl)-iV- [4-(methoxycarbonyl)ben2yl] -N-( 1 - methylpiperidin-4-yl) acetamide;
2-(4-trifluoromethylphenyl)-N-[4-(methoxycarbonyl)benzyl]-N-( 1 - methylpiperidin-4-yl) acetamide;
2-Phenyl-N-[4-(methoxycarbonyl)benzyl]-iV-( 1 -methylpiperidin-4-yl) acetamide;
2-(4-Chlorophenyl)-N- [4-(methoxycarbonyl)benzyl] -N-( 1 -methylpiperidin- 4-yl) acetamide;
2-(4-Methoxyphenyl)-7V-[4-(methoxycarbonyl)benzyl]-iV-(l- methylpiperidin-4-yl) acetamide;
2 -(4-methoxyphenyl)-N-(2-4(fluorophenyl) ethyl)-N-( 1 -methy lpiperidin-4- yl) acetamide;
2-(4-methoxyphenyl)-iV-[2-(2,5-dimethoxyphenyl)ethyl]-iV-(l-methyl piperidin-4-yl) acetamide;
2-(4-methoxyphenyl)-7V-[2-(2,4-dichlorophenyl) ethyl]-7V-(l - methylpiperidin-4-yl) acetamide;
2-(4-methoxyphenyl)-iV-[2-(3-chlorophenyl) ethyI]-iV-(l -methylpiperidin- 4-yl) acetamide;
2-(4-methoxyphenyl)-N-[2-(4-methoxyphenyl) ethyl] -N-( 1 - methylpiperidin-4-yl) acetamide;
2-(4-methoxyphenyl)-N-[2-(3-fluorophenyl) ethyl]-N-(l -methylpiperidin- 4-yl) acetamide;
2-(4-ethoxyphenyl)-N-[2-(4-fluorophenethyl]-N-(l-methylpiperidin-4-yl) acetamide;
2-(4-ethoxyphenyl)-iV:-(4-fluoroben2yl)-//-( 1 -methylpiperidin-4-yl) acetamide; 2-(4-Chlorophenyl)-N-(4-methylbenzyl)-N-(l-isopropylpiperidin-4-yl)- acetamide;
2-(4-Chlorophenyl)-N-(4-methylbenzyl)-N-(l-ethylpiperidin-4-yl)- acetamide;
2-Phenyl-N-(4-methylbenzyl)-N-( 1 -methylpiperidin-4-yl)-acetamide;
2-{4-Chlorophenyl)-N-(4-methylbenzyl)-N-(l-cyclopentylpiperidin-4-yl)- acetamide;
2-(4-Fluorophenyl)-N-(4-methylbenzyl)-N-(l-methylpiperidin-4-yl)- acetamide;
2-(4-Chlorophenyl)-N-(4-methylbenzyl)-N-(l-(2-hydroxyethyl)-piperidin- 4-yl)-acetamide;
2-(4-Chlorophenyl)-N-(4-methylbenzyl)-N-(l-cyclobutylpiperidin-4-yl)- acetamide;
2-(4-Methoxyphenyl)-N-(4-methylbenzyl)-N-(l-cyclobutylpiperidin-4-yl)- acetamide;
N-(4-Methylbenzyl)-N-( 1 -methylpiperidin-4-yl)-N '-benzyl-carbamide;
2-Phenyl-N-(4-methoxybenzyl)-N-(l-methylpiperidin-4-yl)-acetamide;
2-(4-Trifluoromethylphenyl)-N-(4-methoxybenzyl)-N-(l-methylpiperidin- 4-yl)-acetamide;
2-(4-Fluorophenyl)-N-(4-methoxyben2yl)-N-(l-methylpiperidin-4-yl)- acetamide;
2-(4-Methoxyphenyl)-N-(4-methoxybenzyl)-N-(l-methylpiperidin-4-yl)- acetamide;
2-(4-Methylphenyl)-N-(4-chloroben2yl)-N-(l-methylpiperidin-4-yl)- acetamide;
2-(4-Hydroxyphenyl)-N-(4-methylbenzyl)-N-(l-methylpiperidin-4-yl)- acetamide;
2-(4-Methoxyphenyl)-2,2-ethylene-N-(4-methylbenzyl)-N-( 1 - methyIpiperidin-4-yl) acetamide;
2-(4-Methoxyphenyl)-N-alpha-methylbenzyl-N-( 1 -methylpiperidin-4-yl) acetamide;
N-Phenethyl-N-(4-methylbenzyl)-N-( 1 -methylpiperidin-4-yl)-amine; N-(4-Methylbenzyl)-N-(l -methylpiperidiri-4-yl)-N '-(4-methoxybenzyl)- carbamide;
2-(3 ,4-dimethoxyphenyl)-N-(4-methylbenzyl)-N-( 1 -methylpiperidin-4-yl) acetamide;
2-(3,4-Methylenedioxyphenyl)-N-(4-methylbenzyl)-N-(l-methylpiperidin- 4-yl) acetamide;
2-(4-Methoxyphenyl)-N-(4-methylbenzyl)-N-(l-t-butylpiperidin-4-yl)- acetamide;
N-(4-Methylbenzyl)-N-(l-t-butylpiperidin-4-yl)-N'-(4-methoxybenzyl)- carbamide;
2-(4-Ethoxyphenyl)-N-(4-methylbenzyl)-N-( 1 -methylpiperidin-4-yl) acetamide;
2-(4-Butoxyphenyl)-N-(4-methylbenzyl)-N-( 1 -methylpiperidin-4-yl) acetamide;
2-(4-i-Proρoxyphenyl)-N-(4-methylbenzyl)-N-(l-methylpiρeridin-4-yl) acetamide;
N-(4-Fluorobenyzl)-N-(piperidin-4-yl)-2-(4-isobutoxyphenyl)acetamide;
N-(4-Fluorobenzyl)-N'-(4-isopropoxybenzyl)-N-piperidin-4-yl-carbamide, oxalate;
N-( 1 -Beiizyloxycarbonylpiperidin-4-yl)-N-(4-fluorobenzyl)-N'-(4- isopropoxybenzyl)carbamide;
2-(4-methoxyphenyl)-N-(3-phenyl-l-propyl)-N-(l-methylpiperidin-4-yl) acetamide;
2-(4-methoxyphenyl)-N-[2-(4-methylphenyl)ethyl] -N-( 1 -methylpiperidin- 4-yl) acetamide;
2-(4-methoxyphenyl)-N-[2-(4-nitrophenyl)ethyl]-N-(l-methylpiperidin-4- yl) acetamide;
2-(4-methoxyphenyl)-N-(4-methylbenzyl)-N-(l-cyclopentylpiperidin-4-yl) acetamide;
N-((4-(hydroxymethyl)phenyl)methyl)-N-(l-methylpiperidin-4-yl)-2-(4- methoxyphenyl) acetamide;
N-((4-methylphenyl)methyl)-N-(l-methylpiperidin-4-yl)-2-(3-hydroxyl-4- methoxyphenyl) acetamide; N-((4-methylphenyl)methyl)-N-(l-methylpiperidin-4-yl)-2-(3,4- dihydroxyphenyl) acetamide;
N-((3-hydroxy-4-methylphenyl)methyl)-N-(l-methylpiperidin-4-yl)-2-(4- methoxyphenyl) acetamide;
N-((4-methylphenyl)methyl)-N-(l-methylpiperidin-4-yl)-2-(4- bromophenyl) acetamide;
N-((4-methylphenyl)methyl)-N-(l-methylpiperidin-4-yl)-2-(4-iodophenyl) acetamide;
N-((4-methylphenyl)methyl)-N-(l-methylpiperidin-4-yl)-2-(4-(2- propyl)phenyl) acetamide;
N-((4-methylphenyl)methyl)-N-( 1 -methylpiperidin-4-yl)-2-(4- trifluoromethoxyphenyl) acetamide;
N-((4-methylphenyl)methyl)-N-( 1 -methylpiperidin-4-yl)-2-(4- methylthiophenyl) acetamide;
N-((4-methylphenyl)methyl)-N-(l-methylpiperidin-4-yl)-2-(4-(N,N'- dimethylamino)phenyl) acetamide;
N-((4-methylphenyl)methyl)-N-(l-methylpiperidin-4-yl)-2-(4-nitrophenyl) acetamide
N-((4-methylpheny l)methyl)-N-( 1 -methylpiperidin-4-y l)-2-(4-methoxy-3 - methylphenyl) acetamide;
N-((4-methylphenyl)methyl)-N-(l-methylpiperidin-4-yl)-2-(4-pyτidyl) acetamide;
N-((4-methylphenyl)methyl)-N-( 1 -methylpiperidin-4-yl)-2-(4- methylphenyl) acetamide;
2-(4-methoxyphenyl)-N-(phenylethyl)-N-( 1 -methylpiperidin-4-yl) acetamide;
N-((4-methylphenyl)methy I)-N-(I -(phenylmethyl)piperidin-4-yl)- N '- phenylmethylcarbamide;
N-((4-methylphenyl)methy I)-N-(I -(phenylmethyl)piperidin-4-yl)- N '- phenylmethylcarbamide;
N-((4-methylphenyl)methyl)-N-(l-(phenylmethyl)piperidin-4-yl)-4- methoxyphenylacetamide; N-((4-methylphenyl)methyl)-N-( 1 -(phenylmethyl)piperidin-4-yl)-4- methoxyphenylacetamide ;
2-(4-methoxyphenyl)-N-(4-methylbenzyl)-N-( 1 - cyclohexylmethylpiperidin-4-yl) acetamide;
N-((4-methylphenyl)methyl)-N-( 1 -(phenylmethyl)piperidin-4-yl)-4~ methoxyphenylthioacetamide;
2-(4-Methoxyphenyl)-N-(4-methylbenzyl)-N-[ 1 -(2-methylthiazol-4- ylmethyl) piperidin-4-yl] acetamide;
2-(4-Methoxyphenyl)-jV-[2-(2-thienyl)ethyl]-N-(l-rnethylpiperidin-4-yl) acetamide;
2-(4-Methoxyphenyl)-N-(2-thienylmethyl)-iV-(l-methylpiperidin-4-yl) acetamide;
2-(4-Methoxyphenyl)-iV-(furfuryl)-N-( 1 -methylpiperidin-4-yl) acetamide;
2(2-thienyl)-iV-(4-methylphenylmethyl)-N-(l-methylpiperidin-4-yI) acetamide;
N-[ 1 -((S)-3 ,5-Dihydroxypentyl)piperidine-4-yl]-N-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)acetamide, tartrate; and
N-[l-((R)-3J5-Dihydroxypentyl)piperidine-4-yl]-N-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)acetamide, tartrate.
{0085] In some embodiments, the compound of Formula I is selected from the group consisting of:
N-( 1 -((2-bromophenyl)methyl)piperidin-4-yl)-iV-((4- methylphenyl)methyl)-jV-phenylmethylcarbamide;
Λr-(l-((4-hydroxy-3-methoxyphenyl)methyl)piperidin-4-yl)-7Vr-((4- methylphenyl)methyl)--V-phenylmethylcarbamide;
N-( 1 -((5-ethylthien-2-y l)methyl)piperidin-4-yl)-N-((4- methylphenyl)methyl)-N'-phenylmethylcarbamide;
N-(l-(imidazol-2-ylmethyl)piperidin-4-yl)-N-((4-methylphenyl)methyl)- N'-phenylmethylcarbamide;
N-( 1 -((4-fluorophenyl)methyl)piperidin-4-yl)-N-((4- methylphenyl)methyl)-N'-phenylmethylcarbamide;
N-((4-methylphenyl)methyl)-N-( 1 -(phenylmethyl)piperdin-4-yl)-N'- phenylmethylcarbamide; N-((4-methylphenyl)methyl)-N-( 1 -(phenylmethy l)piperdin-4-yl)-4- methoxyphenylacetamide;
N-((4-methylphenyl)methyl)-N-(l-((4-methylphenyl)methyl)piperidin-4- yl)-4-methoxyphenylacetamide;
N-( 1 -((4-hydroxyphenyl)methyl)piperidin-4-yl)-N-((4- methylphenyl)methyl)-4-methoxyphenylacetamide;
N-( 1 -((2-hydroxyphenyl)methyl)piperidin-4-yl)-N-((4- methylphenyl)methyl)-4-methoxyphenylacetamide;
N-(I -(phenylmethyl)piperidin-4-yl)-N-(3 -phenyl-2-propen- 1 -yl)-4- methoxypheny lacetamide ;
N-((Methylphenyl)methyl)-N-( 1 -(phenylmethyl)piperdin-4-yl)-4- methoxyphenylthioacetamide; and
2-(4-Methoxyphenyl)-N-(4-methylbenzyl)-N-[ 1 -(2-methylthiazol-4- ylmethyl)piperdin-4-yl] acetamide.
[0086] In some embodiments, the compound of Formula I is selected from the group consisting of:
N-((4-methylphenyl)methyl)-N-(l-(phenylmethyl)pyrrolidin-3-yl)-N'- phenylmethylcarbamide; and
N-((4-methylphenyl)methyl)-N-(l-(phenylmethyl)pyrrolidin-3-yl)-4- methoxyphenylacetamide.
[0087] In one embodiment, the compound of Formula I is 2-(4- methoxyphenyl)-N-(4-methylbenzyl)-N-(8-methyl-8-aza-bicyclo[3.2.1 ]oct-3-yl)- acetamide.
[0088] In some embodiments, the compound of Formula I is selected from the group consisting of:
N-( 1 -( 1 -methylethyl)piperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide;
N-(l-(2,2-dimethylethyl)ρiperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide;
N-(I -pentylpiperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide;
N-( 1 -hexylpiperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide; N-( 1 -cyclohexylpϊperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetam.de;
N-( 1 -cyclopenty lpiperidin-4-y l)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide;
N-( 1 -cyclobutylpiperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide ;
N-(cyclopropylpiperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide;
N-(l-(cyclopentylmethyl)piperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide;
N-(l-(cyclobutylmethyl)piperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide;
N-(l-(cyclopropylmethyl)piperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide ;
N-(l-(2-hydroxyethyl)piperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide;
N-(l-(3-hydroxyethyl)piperidin-4-yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide;
N-((4-methylphenyl)methyl)-N-( 1 -methylipieridin-4-yl)-4- methoxyphenylacetamide;
N-(l-ethylpiperidin-4yl)-N-((4-methylphenyl)methyl)-4- methoxyphenylacetamide ;
N-((4-methylphenyl)methyl)-N-( 1 -propylpiperidin-4-yl)-4- methoxyphenylacetamide ;
N-(I -butylpiperidin-4-yl)-N-((4-methylphenyl)methyI)-4- methoxyphenylacetamide;
2-Phenyl-N-[4-(methoxycarbonyl)benzyl]-N-(l-methylpiperidin-4- yl)acetamide;
2-(4-Chlorophenyl)-N-[4-(methoxycarbonyl)benzyl]-N-(l- methylpiperidin-4-yl)acetamide ;
2-(4-methoxyphenyl)-N-(4-methylbenzyl)-N-[l-(4-chloromethyl-2- thiazolylmethyl)piperidin-4-yl]acetamide; 2-(4-methoxyphenyl)-N-(4-methylbenzyl)-N-{ 1 -[2-(2- hydroxyethoxy)ethyl]piperidin-4-yl } acetamide;
2-(4-methoxyphenyl)-N-(4-methylbenzyl)-N-[l-((2-chIoro-5- thienyl)methyl)piperidin-4-yl]acetamide;
2-(4-methoxyphenyl)-N-(4-methylbenzyl)-N-{ 1 -[2-(3- indolyl)ethyl]piperidin-4-yl}acetamide;
2-(4-methoxyphenyl)-N-(4-methylbenzyl)-N-{l-[3-(l,2,4-triazol-l- yl)propyl]piperidin-4-yl} acetamide;
2-(4-methoxyphenyl)-N-(4-methylbenzyl)-JV-[l-(5-benzofurazanyl methyl)piperidin-4-yl] acetamide;
2-(4-methoxyphenyl)-N-(4-methylbenzyl)-N-[l-(5- chlorobenzo[b]thien-3-ylmethyl)piperidin-4-yl] acetamide;
2-(4-methoxyphenyl)-N-(4-methylben2yl)-N-[l-(5-phenyl-l,2,4-oxadiazol- 3-ylmethyl)piperidin-4-yl]acetamide;
2-(4-t-Butoxyphenyl)-N-(4-methylbenzyl)-N-( 1 -methylpiperidin-4- yl)acetamide;
2-(4-Butoxyphenyl)-N-(4-fluoroben2yl)-N-( 1 -methyIpiperidin-4- yl)acetamide;
2-(4-Propoxyphenyl)-N-(4-flourobenzyl)-N-( 1 -methylpiperidin-4- yl)acetamiide;
2~(4-i-Propoxyphenyl)-N-(4-fluorobenzyl)-N-( 1 -rnethylpiperidin-4- yl)acetamide; and
2-(4-t-Butoxyphenyl)-N-(4-fluorobenzyl)-N-( 1 -methylpiperidin-4- yl)acetamide.
[0089] In some embodiments, the compound of Formula I is selected from the group consisting of:
N-(4-Methylbenzyl)-N-(l-methylpiperidin-4-yl)-N'-phenyl-carbamide;
N-Phenethyl-N-(l-methylpiperidin-4-yl)-N'-benzyl-carbamide;
N-Phenethyl-N-( 1 -methylpiperidin-4-yl)-N '-phenyl-carbamide;
N-(3-Phenylpropyl)-N-(l-methylpiperidin-4-yl)-N'-benzyl-carbamide;
N-(3-Phenylpropyl)-N-(l-methylpiperidin-4-yl)-N'-phenyl-carbamide;
2-Phenyl-2-ethyl-N-(4-methylbenzyl)-N-( 1 -methylpiperidin-4-yl) acetamide; N-(4-Methylbenzyl)-N-(l-methylpiperidin-4-yl)-N'-phenethyl-carbamide; and
N-Phenethyl-N-(l-methylpiperidin-4-yl)-N'-phenethyl-carbamide. [0090] In one embodiment, the compound of Formula I is 2-(4- Methoxyphenyl)-N-(4-methylbenzyl)-N-(8-methyl-8-aza-bicyclo[3.2.1]octen-3-yl)- acetamide.
[0091] Other embodiments includes use of compounds of the general Formula (II):
Figure imgf000031_0001
or a pharmaceutically acceptable salt, amide, ester, or prodrug thereof, where: Zi is
Figure imgf000031_0002
R1 is selected from the group consisting of optionally substituted heterocyclyl, and optionally substituted (heterocyclyl)Ci-6-alkyl.
R2 and R3 are independently selected from the group consisting of hydrogen, Ci-6-alkyl and halogen or such that R2 together with R3 forms a ring. For example, R2 and R3 together can form a 3-, A-, S-, 6-, or 7-membered ring system with the atoms of the piperidine ring. m1 is selected from the group consisting of 0, 1 , and 2. n1 is selected from the group consisting of 1, 2, and 3;
Aτ3 is an optionally substituted aryl or heteroaryl. For example, the aryl or heteroaryl can be optionally substituted with a substituent selected from the group consisting of Ci-β-alkyl, Ci^-alkoxy, carboxyl, amino, hydroxy, thiol, nitro, cyano, guanidino, carbamido and halogen.
Wi is selected from the group consisting of O and S. X3 is selected from the group consisting of optionally substituted methylene, optionally substituted ethylene, optionally substituted propylene, optionally substituted vinylene, and CH2N(RN1), wherein RN1 is selected from hydrogen and Cj-e-alkyl.
Aτ4 is an optionally substituted aryl or heteroaryl.
[0092] While not being bound by any particular theory, the presence of a heterocyclic substituent at the nitrogen of the piperidine ring of these compounds is believed to improve the bioavailability of the compounds in comparison to related compounds.
[0093] In some embodiments, the heterocyclyl or (heterocyclyl)C]_6-alkyl of R1 may be optionally substituted. The substituent may be selected from halogen, hydroxy, alkyl, alkoxy, and amino. In some embodiments, the substituent may be on the alkyl chain or the ring system. In further embodiments the substituent is on the ring system.
[0094] In certain embodiments, the heterocyclyl ring in R1 may be selected from the group consisting of tetrahydrothiopyran, 4H-pyran, tetrahydropyran, piperidine, 1,3-dioxin, 1,3-dioxane, 1,4-dioxin, 1,4-dioxane, piperazine, 1,3-oxathiane, 1 ,4-oxathiin, 1,4-oxathiane, tetrahydro-l,4-thiazine, 2H-l,2-oxazine, maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, morpholine, trioxane, hexahydro-l,3,5-triazine, tetrahydrothiophene, tetrahydrofuran, pyrroline, pyrrolidine, pyrrolidone, pyrrolidine, pyrazoline, pyrazolidine, imidazoline, ϊmidazolidine, 1,3-dioxole, 1,3-dioxolane, 1,3-dithiole, 1,3-dithiolane, isoxazoline, isoxazolidine, oxazoline, oxazolidine, oxazolidinone, thiazoline, thiazolidine, and 1,3- oxathiolane. In some embodiments, the heterocyclyl ring is selected from 1,3-dioxane, 1,3-dioxolane, and tetrahydropyran.
[0095] The azacyclic ring may be a 5, 6, or 7-membered ring as reflected in that m' may be selected from 0, 1 and 2. In certain embodiments, however, the azacyclic ring is a 6-membered ring, wherein m' is 1.
[0096] The azacyclic ring, further to being substituted at the nitrogen position, may be substituted with R2 and R3. R2 and R3 may be independently selected from the group consisting of hydrogen, Ci-6-alkyl, and halogen, or such that R together with R forms a ring. That is to say that R2 and R3 may be biradicals which combine to form a 3-, 4-, 5-, 6-, or 7-membered ring system with the atoms of the azacyclic ring.
[0097] In some embodiments, the azacyclic ring system is selected from
Figure imgf000033_0001
wherein R7 and R8 are independently selected from the group consisting of hydrogen, halogen, hydroxyl, and Cj. β alkyl. In certain embodiments R7 and R8 are hydrogen.
[0098] In other embodiments, R2 and R3 are hydrogen.
[0099] In some embodiments, R1 is an optionally substituted (heterocyclyl)Ci_ 6-alkyl. In certain of these embodiments, R1 is an optionally substituted (heterocyclyl)methyl, an optionally substituted (heterocyclyl)ethyl, or an optionally substituted (heterocyclyl)propyl. In other embodiments, R1 is an optionally substituted (hetero cycly l)ethyl .
[0100] Ar3 is linked to a central nitrogen atom via a short aliphatic chain 1, 2, or 3 carbon atoms in length. In certain embodiments, n' is 1, resulting in a methylene spacer between the central nitrogen atom and Ar3. Ar3 may be an optionally substituted aryl or heteroaryl. In some embodiments, Ar3 is an optionally substituted aryl. In some embodiments, the central nitrogen atom is linked to an optionally substituted benzyl group.
[0101] In certain embodiments Ar3 is an optionally substituted aryl, which may be a 4-substituted aryl. The 4-substituent of the 4-substituted aryl may be any substituent known to the person skilled in the art, such as a C1 -6-alkyl, Q-6-alkoxy, carboxyl, amino, hydroxy, thiol, nitro, cyano, guanidino, carbamido and halogen. In some embodiments, the halogen is fluoro, while in other embodiments, the halogen is chloro.
[0102] In other embodiments, Ar3 is selected from the group consisting of alkyl-substituted phenyl, alkoxy-substituted phenyl, halogen-substituted phenyl, hydroxy- substituted phenyl and amino-substituted phenyl. In some embodiments, the substituent may be present 0 to 5 times, or 0 to 4 times, or 0 to 3 times, such as 0, 1 , 2, or 3 times. In certain embodiments, the substituent is present 1 to 2 times. In some embodiments, Ar3 is a 4-substituted aryl selected from the group consisting of 4-halophenyl and 4-alkylphenyl. In some embodiments, the phenyl group is 4-fluorophenyl.
[0103] In other embodiments, Ar3 is an optionally substituted heteroaryl. The heteroaryl may be substituted with substituents known to the person skilled in the art, such as a Ci-g-alkyl, C]_6-alkoxy, carboxyl, amino, hydroxy, thiol, nitro, cyano, guanidino, carbamido and halogen.
[0104] Further to being linked to both the azacyclic ring and to Ar3 via a short aliphatic chain, the central nitrogen is linked to Ar4 via a 2 to 4 carbon spacer unit. This spacer unit comprises a carbonyl or thiocarbonyl function wherein Wi is selected from the group consisting of oxygen and sulfur. In some embodiments Wi is oxygen.
[0105] In certain embodiments, X3 may be selected from the group consisting of optionally substituted methylene, optionally substituted ethylene, optionally substituted propylene, optionally substituted vinylene, and CH2N(R1""). Thus X3 may extend the spacer unit by 1 to 3 atoms between the central nitrogen and Ar4 and render the central nitrogen part of an amide or carbamide. In some embodiments, X3 is selected from the group consisting of optionally substituted methylene, optionally substituted ethylene, and CH2N(R1"11). In some embodiments, X3 is an optionally substituted methylene, or CH2N(RN1), wherein RNl may be hydrogen.
[0106] In certain embodiments, Ar4 may be an optionally substituted aryl or heteroaryl. In certain embodiments, Ar4 is an optionally substituted aryl. In some embodiments, Ai4 is a 4-substituted aryl.
[0107] In a further embodiment, Ar4 may be selected from the group consisting of alkoxy-substituted phenyl, halogen-substituted phenyl, hydroxy-substituted phenyl, amino-substituted phenyl, and heterocyclyl-substituted phenyl.
[0108] In certain embodiments, Ar4 is a 4-substituted aryl wherein the substituent is selected from the group consisting of alkyl, alkoxy, halogen, hydroxy, amino, alkylamino, heterocyclyl, and heteroaryl. In some embodiments, the substituent on Ar4 is selected from chloro, fluoro, hydroxy, methoxy, ethoxy, propoxy, isopropoxy, n- butoxy, sec-butoxy, tert-butoxy, trifluoromethoxy, N-morpholinyl, N-pyrrolidinyl, N- pyrazolyl, N-triazolyl and 2-oxopyrrolidinyl.
[0109] Suitable embodiments of the compounds of Formula II can be selected from the group consisting of: iV-{l-[2-(l,3-Dioxolan-2-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-N'-
(4-isobutoxybenzyl)carbamide, hydrochloride;
N- { 1 -[2-(1 ,3-Dioxan-2-yl)ethyl]piperidin-4-yl } -N-(4-fluorobenzyl)-2-[4-
(2-hydroxy-2-methylpropoxy)phenyl]acetamide, tartrate; iV-{l-[3-(3,5-Dimethylpiperidin-l-yl)propyl]piperidin-4-yl}-N-(4- fluorobenzyl)-2-(4-isobutoxyphenyl)acetamide, dihydrochloride;
1 - [3 -(4- { (4-Fluorobenzyl)-[2-(4-isobutoxyphenyl)acetyl] amino } piperidin- l-yl)propyl]piperidine-4-carboxylic acid methyl ester, dihydrochloride;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{ 1 -[2-(I -methylpyrrolidin-2- yl)ethyl]piperidin-4-yl} acetamide. dioxalate;
N- { 1 -[3-(2,6-Dimethylmorpholin-4-yl)propyl]piperidin-4-yl } -N-(4- fluorobenzyl)-2-(4-isobutoxyphenyl)acetamide5 dioxalate;
N-(4-Fluorobenzyl)-N-{l-[3-(3-hydroxypiperidin-l-yl)propyl]piperidin-4- yl}-2-(4-isobutoxyphenyl)acetamide, dioxalate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{l-[3-(2-methylpiperidin-l- yl)propyl]piperidin-4-yl} acetamide, dioxalate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-[l -(3-pyrrolidin- 1 -yl- propyl)piperidin-4-yl]acetamide, dioxalate;
N-{l-[3-(2,5-Dimethylpyrrolidin-l-yl)propyl]piperidin-4-yl}-N-(4- fluorobenzyl)-2-(4-isobutoxyphenyl)acetamide, dioxalate;
N-(4-Fluorobenzyl)-N-{l-[3-(3-hydroxymethylpiperidin-l- yl)propyl]piperidin-4-yl}-2-(4-isobutoxyphenyl)acetamide, dioxalate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{l-[3-(4-(S)-isopropyl-2- oxo-oxazolidin-3 -yl)propyl]piperidin-4-yl } acetamide, oxalate;
N-[2-(4-Fluorophenyl)ethyl]-2-(4-isobutoxyphenyl)-N-{l-[3-(4-(S)- isoρroρyl-2-oxo-oxazolidin-3 -yl)propyl]piperidin-4-yl} acetamide, oxalate;
N-[2-(4-Fluorophenyl)ethyl]-N-{l-[3-(4-(S)-isopropyl-2-oxo-oxazolidin-3- yI)propyl]piperidin-4-yl}-2-(4-propoxyphenyl)acetamides oxalate;
N-(4-Fluorobenzyl)-N- { 1 -[3-(4-(S)-isopropyl-2-oxo-oxazolidin-3- yl)propyl]piperidin-4-yl } -2-(4-propoxyphenyl)acetamide, oxalate;
N-{l-[2-(l,3-Dioxan-2-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)acetamide, oxalate;
N- { l-[2-(l,3-Dioxan-2-yl)ethyl]piperidin-4-yl}-N-[2-(4- fluorophenyl)ethyl]-2-(4-isobutoxyphenyl)acetamide, oxalate;
N- { 1 -[2-(1 ,3-Dioxan-2-yl)ethyl]piperidin-4-yl} -N-[2-(4- fluorophenyl)ethyl]-2-(4-propoxyphenyl)acetamide, oxalate; N-{l-[2-(l,3-Dioxan-2-yl)ethyl]ρiperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- propoxyphenyl)acetamide, tartrate;
N-{l-[2-(l,3-Dioxan-2-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-N'-(4- isobutoxybenzyl)carbamide, tartrate;
N- { 1 -[2-( 1 ,3-Dioxan-2-yl)ethyl]ρiperidin-4-yl } -N-(4-fluorobenzyl)-2-(4- fluorophenyl)acetamide, tartrate;
N-{l-[2-(l33-Dioxan-2-yl)ethyl]piρeridin-4-yl}-N-(4-fluorobenzyl)-2-p- tolylacetamide, tartrate;
2-Benzofuran-5-yl-N-{l-[2-(l,3-dioxan-2-yl)ethyl]piperidin-4-yl}-N-(4- fluorobenzyl)acetamide, tartrate;
2-(2,3-Dihydrobenzofuran-5-yl)-N-{ 1 -[2-(1 ,3-dioxan-2-yl)ethyl]piperidin- 4-yl } -N-(4-fluorobenzyl)acetamide, tartrate;
N- { 1 -[2-(2,2-Dimethyl- 1 ,3 -dioxolan-4-yl)ethyl]piperidin-4-yl }-N-(4- fluorobenzyl)-2-(4-isobutoxyphenyl)acetamide, tartrate;
N-{l-[2-(l,3-Dioxan-4-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)acetamide5 tartrate;
N-{ 1 -[2-(1 ,3-Dioxan-4-yl)ethyl]piρeridin-4-yl}-N-(4-fluorobenzyl)-2-(4- trifluoromethylphenyl)acetamide, tartrate;
2-(4-Cyanophenyl)-N-{l-[2-(l,3-dioxan-4-yl)ethyl]piperidin-4-yl}-N-(4- fluorobenzyl)acetamide, tartrate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N- { 1 - [2-(2-oxo-imidazolidin- 1 -yl)ethyl]piperidin-4-yl } acetamide, hydrochloride;
2-(4-Methoxyphenyl)-N-(4-methylben2yl)-N- { 1 -[2-(2-oxo-imidazolidin- 1 - yl)ethyl]piperidin-4-yl } acetamide, hydrochloride;
N-(4-Fluorobenzyl)-2-(4-isopropoxyphenyl)-N-{l-[2-(2-oxo-imidazolidin- 1 -yl)ethyl]piperidin-4-yl} acetamide, hydrochloride;
N-(4-Fluoroberi2yl)-2-(4-isopropoxyphenyl)-N-{l-[3-(3-methyl-2-oxo-2,3- dihydro-benzoimidazol-l-yl)propyl]piperidin-4-yl}acetamide; hydrochloride;
N-{ l-[2-(2,4-Dioxo-l,4-dihydro-2H-quinazolin-3-yl)ethyl]piperidin-4-yl}- 2-(4-methoxyphenyl)-N-(4-methylbenzyl)acetamide, hydrochloride;
2-(4-Methoxyphenyl)-N-(4-methylbenzyl)-N-{ 1 -[3-(2-oxo-2,3- dihydrobenzoimidazol- 1 -yl)proρyl]piperidin-4-yl } -acetamide, hydrochloride; N-(4-Fluoroben2yl)-2-(4-isopropoxyphenyl)-N-{ 1 -[4-(2-oxo-2,3- dihydrobenzoimidazol-l-yl)butyl]piperidin-4-yl}acetamide, hydrochloride;
N-{l-[2-(2,4-Dioxo-l,4-dihydro-2H-quinazolin-3-yl)ethyl]piperidin-4-yl}- N-(4-fluorobenzyl)-2-(4-isopropoxyphenyl)acetarnide, hydrochloride;
N-{ 1 -[2-(1 ,3-Dioxolan-2-yl)ethyl]ρiperidin-4-yl}-N-(4-fluorobenzyl)-N'- (4-isopropoxy-benzyl)carbamide, oxalate;
N- { 1 -[2-( 1 ,3-Dioxolan-2-yl)ethyl]piperidin-4-yl]-2-(4-methoxyphenyl)-N- (4-methylbenzyl)acetamide, hydrochloride;
N-{l-[2-(l,3-Dioxolan-2-yl)ethyl]piperidin-4-yl>-N-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)acetamide, hydrochloride;
N-{l-[2-(l,3-Dioxolan-2-yl)ethyl]piperidin-4-yl}-2-(4-isopropoxyphenyl)- N-(4-rnethylbenzyl)acetamide, hydrochloride;
N-{l-[2-(l,3-Dioxolan-2-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- propoxyphenyl)acetamide, tartrate;
N-(4-Fluorobenzyl)-N'-(4-isopropoxybenzyl)-N-{ 1 -[2-((S)-4-methyl-l ,3- dioxolane-2-yl)ethyl]piperidin-4-yl} carbamide, oxalate;
N-(4-Fluorobenzyl)-N'-(4-isopropoxybenzyl)-N-[l-(3-morpholin-4-yl- propyl)piperidin-4-yl]carbamide, oxalate;
2-(4-Methoxyphenyl)-iV-(4-methylbenzyl)-N-[l-(2-moφholin-4- ylethyl)piperidin-4-yl]acetamide, dihydrochloride;
2-(4-Methoxyphenyl)-jV-(4-methylbenzyl)-iV-[l-(3-morpholin-4- ylpropyl)piperidin-4-yl]acetamide, dihydrochloride;
JV-(4-Fluoroben2yl)-2-(4-isobutoxyphenyl)-N-[ 1 -(3 -morpholin-4- ylpropyl)piperidin-4-yl]acetamide, dihydrochloride; τV-(4-Fluorobenzyl)-2-(4-isopropoxyphenyl)-iV-[l-(3-moφholin-4-yl- propyl)piperidin-4-yl]acetamide, dihydrochloride;
N-(4-Fluorobenzyl)-N'-(4-isopropoxybenzyl)-N-[l-(3-piperidin-l-yl- propyl)piperidin-4-yl]carbamide, oxalate;
N-(4-Fluorobenzyl)-N'-(4-isopropoxyben2yl)-N-[l-(3-((S)-4-isopropyl-2- oxazolidinon-1 -yl-propyl)piperidin-4-yl]carbamide, tartrate;
N-(4-Fluorobenzyl)-N'-(4-isopropoxybenzyl)-N-{l-[2-(2,5,5-trimethyl- 1 ,3-dioxan-2-yl)ethyl] } piperidin-4-yl]carbamide, oxalate; N-ll-fS-Cl^-Dioxolan^-yOpropyllpiperidin^-yO-N-C^fluorobenzyO-N'- (4-isopropoxybenzyl)carbamide, oxalate;
N-[l-(2,2-Dimethyl-l,3-dioxan-5-yl)piperidin-4-yl]-N-(4-fluorobenzyl)- N '-(4-isopropoxybenzyl)carbamide, oxalate;
N-(4-Fluorobenzyl)-N ' -(4-isopropoxybenzy I)-N- { [2-( 1 -methyl pyrroli din- 2-yl)ethyl] -piperidin-4-yl } carbamide, oxalate;
N-[l-(2,2-Dimethyl-l53-dioxan-5-yl)piperidin-4-yl]-N-(4-fluorobenzyl)-2- (4-i sobutoxyphenyl)acetamide, oxalate;
N-[I-(1 ,3-Dioxan-5-yl)-piperidin-4-yl)-N-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)acetamide, tartrate;
N- [ 1 -(2,2-Dimethyl- 1 ,3-dioxan-5-yl)piperidin-4-yl]-N-(4-fluorobenzyl)-2- (4-fluorophenyl)acetamide, tartrate;
N-{l-[2-(l,3-Dioxan-4-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- fluorophenyl)acetamide, tartrate:
N-{ l-[2-(l,3-Dioxan-4-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- trifluoromethoxyphenyl)acetamide, tartrate :
N-{ l-[2-(l,3-Dioxan-4-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- propoxyphenyl)acetamide, tartrate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-[ 1 -(tetrahydropyran-4- yl)piperidin-4-yl]acetamide, tartrate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-[l-(tetrahydropyran-4- ylmethyl)piρeridin-4-yl]acetamide, tartrate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{l-[2-(tetrahydropyran-4- yl)ethyl]piperidin-4-yl]acetamide, tartrate;
N-(4-Fluoroben2yl)-2-(4-fluorophenyl)-N- [ 1 -(tetrahydropyran-4- yl)piperidin-4-yl]acetamide, tartrate;
N-{l-[2-((4S)-l,3-Dioxane-4-yl)ethyl]piperidine-4-yl}-N-(4- fluorobenzyl)-2-(4-isobutoxyphenyl)acetamide, tartrate;
2-(4-Benzyloxyphenyl)-N-{l-[2-(l,3-dioxan-2-yl)ethyl]piperidin-4-yl}-N- (4-fluorobenzyl)acetamide, tartrate;
N-{ 1 -[2-(I >3-Dioxan-2-yl)ethyl]ρiperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- hydroxyphenyl)-acetamide, tartrate; N- { 1 -[2-( 153-Dioxan-2-yl)ethyl]piperidin-4-yl } -N-(4-fluorobenzyl)-2-(4- methoxyphenyl)-acetamide, tartrate;
N-{l-[2-(1.3-Dioxan-2-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- isopropylphenyl)-acetamide, tartrate;
N- { 1 -[2-( 1 ,3-Dioxan-2-yl)ethyl]piρeridin-4-yl} -N-(4-fluorobenzyl)-2-(4- trifluoromethoxy-phenyOacetamide, tartrate;
N-{l-[2-(l33-Dioxan-2-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- ethoxyphenyl)-acetamide, oxalate;
N-{l-[2-(l53-Dioxan-2-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- isopropoxyphenyl)-acetamide, oxalate;
N-{l-[2-(l,3-Dioxan-2-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2- phenylacetamide, oxalate;
N- { 1 -[2-(1 ,3-Dioxan-2-yl)ethyl]piperidin-4-yl} -N-(4-fluorobenzyl)-2-[4- (2-fluoroethoxy)-phenyl]acetamide, oxalate;
N- { 1 -[2-(5 ,5-Dimethyl- 1 ,3 dioxan-2-yl)ethyl]piperidin-4-yl } -N-(4- fluoroben2yl)-2-(4-isobutoxyphenyl)acetamide, oxalate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N- { 1 - [2-((R)-4-methyl- 1 ,3- dioxan-2-y l)ethy 1] -piperidin-4-yl } acetamide, oxalate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{l-[2-((S)-4-methyl-l,3- dioxolan-2-yl)ethyl]-piperidin-4-yl } acetamide, oxalate;
N-{l-[2-(4,6-Dimethyl-l,3-dioxan-2-yl)ethyl]piperidin-4-yl}-N-(4- fluorobenzyl)-2-(4-isobutoxyρhenyl)acetamide, oxalate;
N-(4-Fluorobenzyl)-N-{ 1 -[2-((S)-4-methyl- 1 ,3-dioxolan-2-yl)ethyl] piperidin-4-yl } -2-(4-trifluoromethoxyphenyl)acetamide, oxalate;
N-(4-Fluorobenzyl)-2-(4-isopropylphenyl)-N-{ 1 -[2-((S)-4-methyl- 1 ,3- dioxolan-2-yl)ethyl]-piperidin-4-yl}acetamide, oxalate; .
N-(4-Fluorobenzyl)-N-{ l-[2-((R)-4-methyl-l ,3-dioxan-2-yl)ethyl] piperidin-4-yl } -2-(4-trifluoromethoxyphenyl)acetamide, oxalate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{ 1 -[2-(255,5-trimethyl- 1 ,3- dioxan-2-yl)ethyl] piperidin-4-yl} acetamide, oxalate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{l-[2-(2-methyl-l,3- dioxolan-2-yl)ethyl]-piperidin-4-yl}acetamide, oxalate; N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{ 1 -[3-(1 ,3-dioxolan-2- yl)propyl]piperidin-4-yl}acetamide, tartrate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{ 1 -(3-piperidin-l-yl- propyl)piperidin-4-yl} -acetamide, dihydrochloride;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{ 1 -[2-(tetrahydropyran-2- yloxy)ethyl]-piperidin-4-yl} acetamide, oxalate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{l-[3-(2-oxo-piperidin-l- yl)propyl]piperidin-4-yl}acetamide; iSf-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{ 1 -[3-(2-oxo-pyrrolidin- 1 - yl)propyl]piperidin-4-yl } acetamide, hydrochloride;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{l-[3-((R)-4-isopropyl-2- oxo-oxazolidin-3-yl)propyl]piperidin-4-yl}acetamide, oxalate;
N-(4-Fluoroben2yl)-2-(4-isobutoxyphenyl)-N- { 1 - [3-(2-oxo-oxazolidin-3 - yl)propyl]piperidin-4-yl } acetamide, oxalate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{l-[3-((S)-4-methyl-2-oxo- oxazolidin-3-yl)propyl]piperidin-4-yl } acetamide, tartrate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{l-[3-((S)-4-ethyl-2-oxo- oxazolidin-3-yl)-propyl]piperidin-4-yl}acetamide, oxalate;
N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyl)-N-{l-[2-(l,3-oxothiolan-2- yl)ethyl]piperidin-4-yl}acetamide, L-tartrate;
2-(4-Bromophenyl)-N-{ l-[2-(l)3-dioxan-2-yl)ethyl)piperidin-4-yl}-N-(4- fluorobenzyl)-acetamide, L-tartrate;
N-{l-[2-(l,3-Dioxan-2-yl)ethyl)piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- isobutylamino-phenyl)acetamide, L-tartrate;
N-{ 1 -[2-(1 ,3-Dioxan-2-yl)ethyl)piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- propylamino-phenyl)acetamide, L-tartrate;
N-{l-[2-(l,3-Dioxan-2-yl)ethyl)piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- (1 -nitropropyl)-phenyl)acetamide5 L-tartrate;
N-{l-[2-(l,3-Dioxan-2-yl)ethyl)piperidin-4-yl}-N-(4-fluorobenzyl)-2-[4- (2-oxopyrrolidin-l -yl)phenyl)acetamide, L-tartrate;
N-{l-[2-(l,3-Dioxan-2-yl)ethyl)piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- isobutylsulfanyl-phenyl)acetamide, L-tartrate; N-{l-[2-(l,3-Dioxan-2-yl)ethyl)piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- iodophenyl)-acetamide, L-tartrate;
2-(4-Acetophenyl)-N-{l-[2-(l,3-dioxan-2-yl)ethyl)piperidin-4-yl}-N-(4- fluorobenzyl)-acetamide, L-tartrate;
2-[4-(l -Hydroxyiminoethyl)phenyl]-N-{ 1 -[2-(l,3-dioxan-2- yI)ethyl)piperidin-4-yl } -N-(4-fluorobenzyl)acetamide, L-tartrate;
N-{l-[2-(l,3-Dioxan-2-yl)ethyl)piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- moφholin-4-yl-phenyl)acetamide, L-tartrate;
N- { 1 -[2-( 1 ,3-Dioxan-2-yl)ethyl)piperidin-4-yl } -N-(4-fluorobenzyl)-2-(4- pyrazol-l-yl-phenyl)acetamide, L-tartrate;
N-{l-[2-(l,3-Dioxan-2-yl)-l-methylethyl]piperidin-4-yl}-N-(4- fluorobenzyl)-2-(4-iso-butoxyphenyl)-acetamide, L-tartrate;
N-{ l-[2-(l,3-Dioxan-4-yl)ethyl)piperidin-4-yl}-N-(4-fluoroben2yl)-2-(4- pyrazol-1 -yl-phenyl)acetamide, L-tartrate;
N- { 1 -[2-((4R)- 1 ,3-Dioxane-4-yl)ethyl]piperidine-4-yl } -N-(4- fluorobenzyl)-2-(4-isobutoxyphenyl)acetamide, tartrate;
N-{ l-[2-(ls3-Dioxan-2-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-[4- (1 ,2,4-triazol-4-yl)phenyl]acetamide, L-tartrate;
2-(4-methoxyphenyl)-N-(4-methylben2yl)-N-{ 1 -[2-( 1 ,3-dioxolan-2- yl)ethyl]piperidin 4-yl} acetamide;
2-(4-methoxyphenyl)-iV-(4-methylbenzyI)-iV-{l-[3(l,3-dihydro-2H- benzimidazol-2-on-l-yl) propyl] piperidin-4-yl} acetamide;
2-(4-methoxyphenyl)-N-(4-methylbenzyl)-Λ'-[l-(2-(imidazolidinon-l- yl)ethyl)piperidin-4-yl] acetamide;
2-(4-methoxyphenyl)-iV-(4-methylbenzyl)-N- { 1 -[2-(2,4( 1 H,3H) quinazolinedion-3-yl)ethyl] piperidin-4-yl} acetamide;
2-(4-methoxyphenyl)-N-(4-methylbenyl)-N-{ 1 -[2-(1 ,3-dioxolan-2- yl)ethyl]piperidin-4-yl } acetamide;
2-(4-methoxypbenyl)-N-(4-methylbenzyl)-N-[l-(2-(imidazolidinon-l- yl)ethyl)ρiperidin-4-yl]acetamide; and
2-(4-methoxyphenyl)-N-(4-methylbenzyl)-N- { 1 -[2- (2,4(1 H,3H)quinazolinedion-3-yl)ethyl]piperidin-4-yl}acetamide. [0110J Still another embodiment disclosed herein includes use of compounds eneral Formula III:
Figure imgf000042_0001
or a pharmaceutically acceptable salt, amide, ester, or prodrug thereof, wherein:
X4 is selected from the group consisting of CH2, CH2CH2, CH2O, OCH2, O, CH2S5 SCH2, S, CH2N(RN2), N(RN2)CH2 and N(RN2); wherein RN2 is selected from hydrogen and C 1.6 alkyl.
W2 is selected from the group consisting of O and S.
Z3 is absent or selected from the group consisting of CH and N.
R9 is hydrogen, or an optionally substituted substituent selected from the group consisting of Q-e alkyl, C2-8 alkenyl, C2-S alkynyl, C3.8 cycloalkyl, aryl, heteroaryl, aryl(Ci^ alkyl), heteroaryl(Ci.6 alkyl), heterocyclylCCj-β alkyl), C3.8 cyc!oalkyl(Ci.6 alkyl), hydroxy(C].6 alkyl), amino(Ci.6 alkyl), and halo(C1.6 alkyl). m" is selected from the group consisting of 0 and 1.
R12, R13, and R14 are independently hydrogen, or an optionally substituted substituent selected from the group consisting of Ci-6 alkyl, aryl(C|.6 alkyl), heteroarylCd-β alkyl), heterocyclyl(Ci.6 alkyl), hydroxy(Ci-6 alkyl), amino(Ci.6 alkyl), halo(Ci.6 alkyl), C3-6 cycloalkyl, aryl. and heteroaryl, wherein at least two of R12, R13, and R14are independently selected from the group consisting of aryl(C|.6 alkyl), heteroaryl(C 1.6 alkyl), and heterocyclyl(C|.6 alkyl).
R10 and R11 are independently selected from the group consisting of hydrogen, halogen, hydroxy, and optionally substituted C 1.6 alkyl or selected such that R10 and R1 ' together form a ring system such that
Figure imgf000043_0001
is selected from the group consisting of
Figure imgf000043_0002
wherein R15 and R16 are independently selected from the group consisting of hydrogen, halogen, hydroxy, and Cj.6 alkyl.
[0111] Within this embodiment, at least two of R12, R13, and R14 may be independently selected from the group consisting of 4-monosubstituted-aryl(Ci-6 alkyl), and 4- monosubstituted-heteroaryl(Ci.6 alkyl).
[0112] Typically, at least one of the at least two of R12, R13, and R14 independently selected from the group consisting of aryl(Ci-6 alkyl) and heteroaryl(Ci.6 alkyl) is selected from the group consisting of fluoro-substituted-aryl(Ci-6 alkyl), and fiuoro-substituted-heteroaryl(Ci.6 alkyl). Also typically, the other of the at least two of R12, R13, and R14 independently selected from the group consisting of aryl(Ci-6 alkyl) and heteroaryl(Ci_6 alkyl) is selected from the group consisting of (O-Ci-6 alkyl)-substituted- aryl(C i-6 alkyl), and (O-Ci^alkyl)-substituted-heteroaryl(Ci-6 alkyl).
[0113] In some embodiments, at least one of R12, R13, and R14 is selected from the group consisting of fluoro-substituted-aryl(Ci-6 alkyl), and fluoro-substituted- heteroaryl(Ci-6 alkyl).
[0114] Typically, at least two of R12, R13, and R14 are independently selected from the group consisting of aryl(Ci-6 alkyl), heteroaryl(Ci-6 alkyl), and heterocycIyl(Ci-6 alkyl) are each substituted 1, 2, or 3 times, with a substituent selected from the group consisting of halogen and optionally substituted O-CLβ-alkyl. In one aspect, the halogen is fluorine.- In one embodiment, the ring system of one of the at least two of R12, R13, and R14 independently selected from the group consisting of 4UyI(C1-6 alkyl), heteroaryl(C|-6 alkyl), heterocyclyl(Ci_6 alkyl) is substituted 1 to 3 times, such as 1, 2, or 3 times with an optionally substituted O-Ci-6-alkyl, such as a fluorinated O-Ci^-alkyl.
[0115] In yet another embodiment, at least two of R12, RI3 5 and R14 are optionally substituted aryl(Ci.6 alkyl). In a preferred embodiment, at least two of R12, R13, and R14 are optionally substituted benzyl.
[0116] As stated, at least two of R12, R13, and R14 are independently selected from the group consisting of aryl(Ci-6 alkyl), heteroary^Cμβ alkyl), heterocyclyl(Ci.6 alkyl). Typically, the d.6 alkyl of the aryl(Ci_6 alkyl), heteroaryl(C1-6 alkyl), heterocyclyl(Ci-6 alkyl) is Ci_4 alkyl, such as methylene (Ci alkyl). ethylene (C2 alkyl), or propylene (C3 alkyl), or butylene (C4 alkyl), more typically a Cj alkyl or C2 alkyl, most typically a C1 alkyl. In a suitable embodiment, the Ci-6 alkyl of the aryl(Ci-6 alkyl), heteroaryl(Cj.6alkyl), heterocycly^C^ alkyl) may be substituted so as to form a branched hydrocarbon.
[0117] In a combination of embodiments, at least two of R12, R13, and R14 are an optionally substituted benzyl. One of R12, R13, and R14 may be a 4-halo-benzyl group and one may be a 4-alkoxy-benzyl group. The 4-halo-benzyl group is typically a 4-fluoro- benzyl. The 4-alkoxy-benzyl group is typically a C2-5 alkoxybenzyl or optionally fluorinated 4-methoxy-benzyl group such as a fluoromethoxy-benzyl, difluoromethoxy- benzyl, trifluoromethoxy-benzyl group, and 2,2,2-trifluorethoxy-benzyl.
[0118] As stated, the compounds of the invention may be selected from the group consisting of (i) l-oxa-4,9-diaza-spiro[5.5]undecan-3-one; (ii) l-oxa-3,8-diaza- spiro[4.5]decan-2-one; (iii) l,3,8-triaza-spiro[4.5]decan-2-one; (iv) 1,2,9-triaza- spiro[5.5]undecan-3-one; (v) l,2,8-triaza-spiro[4.5]decan-3-one; (vi) 1,2, 8-triaza- spiro[4.5]decan-3-one (vii) l,2,4,8-tetraaza-spiro[4.5]decan-3-one; (viii)2,4,9-triaza- spiro[5.5]undecan-3-one; (ix) 2,8-diaza-spiro[4.5]decan-3-one (x) 2-oxa-4,9-diaza- spiro[5.5]undecan-3-one; (xi) l-thia-3,8-diaza-spiro[4.5]decan-2-one; and (xii) l-oxa-3,9- diaza-spiro[5.5]undecan-2-one.
[0119] Suitable embodiments of the compounds of Formula III may be selected from the group consisting of:
4-(4-Fluorobenzyl)-3-(4-methoxybenzyl)-8-methyl- 1 -oxa-3,8-diaza- spiro[4.5]decan-2-one;
3-(4-Ethoxybenzyl)-4-(4-fluorobenzyl)-8-methyl- 1 -oxa-3,8-diaza- spiro[4.5]decan-2-one; 4-(4-Fluorobenzyl)-8-methyl-3-(4-propoxybenzyl)-l-oxa-3,8-diaza- spiro[4.5]decan-2-one;
3-(4-Cyclopropylmethoxybenzyl)-4-(4-fluorobenzyl)-8-methyl- 1 -oxa-3 ,8- diaza-spiro[4.5]decan-2-one;
4-(4-Fluorobenzyl)-3 -(4-isopropoxybenzyl)-8-methyl- 1 -oxa-3 ,8-diaza- spiro[4.5]decan-2-one;
3-(4-Butoxybenzyl)-4-(4-fluorobenzyl)-8-methyl- 1 -oxa-3,8-diaza- spiro[4.5]decan-2-one;
4-(4-Fluoroben2yl)-3-(4-isobutoxybenzyl)-8-methyl-l-oxa-3,8-diaza- spiro[4.5]decan-2-one;
3-(4-Difluoromethoxybenzyl)-4-(4-fluorobenzyl)-8-niethyl-l-oxa-3,8- diaza-spiro[4.5]decan-2-one;
4-(4-Fluorobenzyl)-8-methyl-3 -(4-trifluoromethoxybenzyl)- 1 -oxa-3 , 8 - diaza-spiro[4.5]decan-2-one;
4-(4-Flυorobenzyl)-8-methyl-3-(4-pentoxybenzyl)-l-oxa-3,8-diaza- spiro[4.5]decan-2-one;
8-Ethyl-4-(4-fluorobenzyl)-3-(4-isobutoxybenzyl)- 1 -oxa-3, 8-diaza- spiro [4.5]decan-2-one;
4-(4-Fluorobenzyl)-3-(4-isobutoxyben2ryl)-8-isopropyl-l-oxa-3,8-diaza- spiro [4.5]decan-2-one;
8-Cyclopropylmethyl-4-(4-fluorobenzyl)-3-(4-isobutoxybenzyl)-l-oxa-3,8- diaza-spiro[4.5]decan-2-one;
8-Cyclohexylmethyl-4-(4-fluorobenzyl)-3-(4-isobutoxybenzyl)-l-oxa-3,8- diaza-spiro [4.5] decaπ-2-one;
8-Cyclopentyl-4-(4-fluorobenzyl)-3-(4-isobutoxyben2yl)-l-oxa-3,8-diaza- spiro[4.5]decan-2-one;
4-(4-Fluorobenzyl)-3-(4-isobutoxybenzyl)-8-(3-moφholin-4-yl-propyl)-l- oxa-3,8-diaza-spiro[4.5]decan-2-one;
8-(2-[l,3]Dioxolan-2-yl-ethyl)-4-(4-fluorobenzyl)-3-(4-isobutoxybenzyl)- l-oxa-3,8-diaza-spiro[4.5]decan-2-oπe;
4-(4-Fiuorobenzyl)-3-(4-isobutoxybenzyl)-8-[2-(2-oxo-imidazolidin-l-yl)- ethylj- 1 -oxa-3,8-diaza-spiro[4.5]decan-2-one; 4-(4-Fluorobenzyl)-3-(4-isobutoxybenzyl)-8-[3-(2-oxo-2,3-dihydro- benzoimidazol- 1 -yl)-propyl] - 1 -oxa-3 ,8 -diaza-spiro [4.5] decan-2-one;
4-(4-Fluorobenzyl)-3-(4-isobutoxybenzyl)-8-(2-methyl-thiazol-4-yl- methyl)-l-oxa-3,8-diaza-spiro[4.5]decan-2-one;
4-(4-Chlorobenzyl)-3-(4-isobutoxybenzyl)-8-methyl-l-oxa-3,8-diaza- spiro[4.5]decan-2-one;
8-Ethyl-4-(4-chloroben2yl)-3-(4-isobutoxybenzyl)- 1 -oxa-3, 8-diaza- spiro [4.5] decan-2-one;
4-(4-Chlorobenzyl)-3-(4-isobutoxybenzyl)-8-isopropyl-l-oxa-3,8-diaza- spiro[4.5]decan-2-one;
8-Cyclopropylmethyl-4-(4-chlorobenzyl)-3-(4-isobutoxybenzyl)-l-oxa-3,8- diaza-spiro[4.5]decan-2-one; .
8-Cyclohexylmethyl-4-(4-chlorobenzyl)-3 -(4-isobutoxybenzyl)- 1 -oxa-3 ,8- diaza-spiro [4.5] decan-2-one;
8-(2-[l,3]Dioxolan-2-yl-ethyl)-4-(4-chlorobenzyl)-3-(4-isobutoxybenzyl)- 1 -oxa-3, 8-diaza-spiro[4.5]decan-2-one;
4-(4-Chlorobenzyl)-3 -(4-i sobutoxyben2yl)- 8- [2 -(2-oxo-imidazolidin- 1 -yl)- ethyl]-l -oxa-3, 8-diaza-spiro[4.5]decan-2-one;
3-(4-Difluoromethoxybenzyl)-4-(4-fluorobenzyl)-8-methyl- 1 -oxa-3 ,8- diaza-spiro[4.5]decan-2-one;
3-(4-Difluoromethoxybenzyl)-8-ethyl-4-(4-fluorobenzyl)-l-oxa-3,8-diaza- spiro[4.5]decan-2-one;
3-(4-Difluoromethoxybenzyl)-4-(4-fluoroben2yl)-8-isopropyl-l-oxa-3,8- diaza-spiro[4.5]decan-2-one;
8-Cyclopropylmethyl-3-(4-difluoromethoxybenzyl)-4-(4-fluorobenzyl)-l- oxa-3,8-diaza-spiro[4.5]decan-2-one;
8-Cyclohexylmethyl-3-(4-difluoromethoxyben2yl)-4-(4-jfluorobenzyl)-l- oxa-3,8-diaza-spiro[4.5]decan-2-one;
3-(4-Difluoromethoxybenzyl)-8-(2-[ 1 ,3]dioxolan-2-yl-ethyl)-4-(4- fluorobenzyl)-l-oxa-3,8-diaza-spiro[4.5]decan-2-one;
3-(4-Diflυoromethoxybenzyl)-4-(4-fluorobenzyl)-8-[2-(2-oxo- imidazolidin-l-yl)-ethyl]-l-oxa-3,8-diaza-spiro[4.5]decan-2-one; 8-Ethyl-4-(4-fluorobenzyl)-3 -(4-trifluoromethoxybenzyl)- 1 -oxa-3 , 8-diaza- spiro[4.5]decan-2-one;
4-(4-Fluorobenzyl)-8-isoρropyl-3-(4-trifluoromethoxybenzyl)-l-oxa-3.8- diaza-spiro[4.5]decan-2-one;
8-Cyclopropylmethyl-4-(4-fluorobenzyl)-3-(4-trifluoromethoxybenzyl)-l- oxa-3,8-diaza-spiro[4.5]decan-2-one;
8-Cyclohexylmethyl-4-(4-fluorobenzyl)-3-(4-trifluoromethoxybenzyl)-l- oxa-3,8-diaza-spiro[4.5]decan-2-one;
8-Cyclopentyl-4-(4-fluorobenzyl)-3-(4-trifluoromethoxyben2yl)-l-oxa-3,8- diaza-spiro[4.5]decan-2-oπe;
8-(2-[ 1 ,3]Dioxolan-2-yl-ethyl)-4-(4-fluorobenzyl)-3-(4- trifluoromethoxybenzyl)-l-oxa-3,8-diaza-spiro[4.5]decan-2-one;
4-(4-Fluoroben2yl)-8-[2-(2-oxo-imidazolidin-l-yl)-ethyl]-3-(4- trifluoromethoxy ben2yl)-l-oxa-3,8-diaza-spiro[4.5]decan-2-one;
8-Ethyl-4-(4-£luorobenzyl)-3-(4-propoxybenzyl)- 1 -oxa-3 ,8-diaza- spiro[4.5]decan-2-one;
4-(4-Fluorobenzyl)-8-isopropyl-3-(4-propoxybenzyl)-l -oxa-3, 8-diaza- spiro[4.5]decan-2-one;
8-Cyclopropylmethyl-4-(4-fluorobenzyl)-3-(4-propoxyben2yl)-l-oxa-3,8- diaza-spiro[4.5]decan-2-one;
8-Cyclohexylmethyl-4-(4-fluorobenzyl)-3-(4-propoxyben2yl)-l-oxa-3,8- diaza-spiro [4.5] decan-2-one;
8-Cyclopentyl-4-(4-fluorobenzyl)-3-(4-propoxybenzyl)-l-oxa-3,8-diaza- spiro [4.5]decan-2-one;
8-(2-[l,3]Dioxolan-2-yl-ethyl)-4-(4-fluorobenzyl)-3-(4-propoxybenzyl)-l- oxa-3,8-diaza-spiro[4.5]decan-2-one;
4-(4-Fluorobenzyl)-8-[2-(2-oxo-imidazolidin-l-yl)-ethyl]-3-(4- propoxybenzyl)-l-oxa-3,8-diaza-spiro[4.5]decan-2-one;
3-(4-Cyclopropylmethoxybenzyl)-8-ethyl-4-(4-fluorobenzyl)-l-oxa-3,8- diaza-spiro[4.5]decan-2-one;
3-(4-Cyclopropylmethoxybenzyl)-4-(4-fluorobenzyl)-8-isopropyl-l-oxa- 3,8-diaza-spiro[4.5]decan-2-one; 3-(4-Cyclopropylmethoxybenzyl)-8-cyclopropylmethyl-4-(4-fluorobenzyl)- l-oxa-3,8-diaza-spiro[4.5]decan-2-one;
3-(4-Cyclopropylmethoxybenzyl)-8-(2-[l,3]dioxolan-2-yl-ethyl)-4-(4- fluorobenzyl)-l-oxa-3,8-diaza-spiro[4.5]decan-2-one;
3-(4-Cyclopropylmethoxybenzyl)-4-(4-fluorobenzyl)-8-[2-(2-oxo- imidazolidin- 1 -yl)-ethyl] - 1 -oxa-3 , 8-diaza-spiro [4.5] decan-2-one;
8-(2-[l .3]-Dioxan-2-yl-ethyl)-4-(4-fluorobenzyl)-3-(4-isobutoxybenzyl)- 1 - oxa-3,8-diaza-spiro[4.5]decane-2-one;
4-(4-Fluorobenzyl)-3-(4-isobutoxybenzyl)-8-{3-[(S)-4-isopropyl-2-oxo- oxazolidin-3-yl]-propyl}-l-oxa-3,8-diaza-spiro[4.5]decane-2-one;
3-(4-Methoxybenzyl)-8-methyl-4-(4-methylbenzyl)- 1 -oxa-3,8-diaza- spiro [4.5]decan-2-one;
8-Methyl-4-(4-methylbenzyl)-3-(4-trifluoromethoxybenzyl)-l-oxa-3,8- diaza-spiro[4.5]decan-2-one;
1 -(4-Fluorobenzyl)-2-(4-methoxybenzyl)-8-methyl-l ,2,8-triaza- spiro[4.5]decan-3-one;
2-(4-Ethoxybenzyl)- 1 -(4-fluorobenzyl)-8-methyl- 1 ,2,8-triaza- spiro[4.5]decan-3-one; l-(4-Fluoroben2yl)-8-methyl-2-(4-propoxybenzyl)-l,2,8-triaza- spiro[4.5]decan-3-one;
1 -(4-Fluorobenzyl)-2-(4-isopropoxybenzyl)-8-methyl-l ,2,8-triaza- spiro[4.5]decan-3-one;
2-(4-Butoxybenzyl)-l-(4-fluorobenzyl)-8-methyl-l,2,8-triaza- spiro[4.5]decan-3-one;
2-(4-Cyclopropylmethoxybenzyl)-l -(4-fluorobenzyl)-8-methyl- 1 ,2,8- triaza-spiro[4.5]decan-3-one;
1 -(4-Fluorobenzyl)-2-(4-isobutoxybenzyl)-8-methyl-l ,2,8-triaza- spiro[4.5]decan-3-one;
2-(4-Difluoromethoxybenzyl)-l-(4-fluorobenzyl)-8-methyl-l,2,8-triaza- spiro [4.5] decan-3 -one;
1 -(4-Fluorobenzyl)-8-methyl-2-(4-trifluoromethoxyben2yl)- 1 ,2,8-triaza- spiro[4.5]decan-3-one; l-(4-Fluorobenzyl)-8-methyl-2-(4-pentoxybenzyl)-l,2,8-triaza- spiro[4.5]decan-3-one;
1 -(4-Chlorobenzyl)-2-(4-ethoxybenzyl)-8-methyl- 1 ,2,8-triaza- spiro[4.5]decan-3-one;
1 -(4-Chloroben2yl)-8-methyl-2-(4-propoxybenzyl)- 1 ,2,8-triaza- spiro[4.5]decan-3-one;
1 -(4-Chlσrobenzyl)-2-(4-isobutoxybenzyl)-8-methyl- 1 ,2,8-triaza- spiro[4.5]decan-3-one;
1 -(4-Chlorobenzyl)-2-(4-cyclopropylmethoxybenzyl)-8-methyl- 1 ,2,8- triaza-spiro[4.5]decan-3-one; l-(4-Chlorobenzyl)-2-(4-difluoromethoxybenzyl)-8-methyl-l,2,8-triaza- spiro[4.5]decan-3-one;
1 -(4-Ethylbenzyl)-2-(4-ethoxybenzyl)-8-methyl-l ,2,8-triaza- spiro[4.5]decan-3-one; l-(4-Ethylbenzyl)-2-(4-isopropoxybenzyl)-8-methyl-l ,2,8-triaza- spiro[4.5]decan-3-one;
1 -(4-Ethylbenzyl)-2-(4-isobutoxybenzyl)-8-methyl- 1 ,2, 8-triaza- spiro [4.5] decan-3 -one;
1 -(4-Ethylbenzyl)-2-(4-cyclopropylmethoxybenzyl)-8-methyl- 1 ,2,8-triaza- spiro [4.5] decan-3 -one;
1 -(4-Ethylbenzyl)-8-methyl-2-(4-trifluoromethoxybenzyl)- 1 ,2,8-triaza- spiro[4.5]decan-3-one;
2-(4-Difluoromethoxybenzyl)-l -(4-fluorobenzyl)-8-ethyl- 1 ,2,8-triaza- spiro[4.5]decan-3-one;
2-(4-Difluoromethoxybenzyl)-l-(4-fluorobenzyl)-8-isopropyl-l,2,8-triaza- spirσ[4.5]decan-3-one;
2-(4-Difluoromethoxybenzyl)-l-(4-fluoroben2yl)-8-cyclopropylmethyl- l,2,8-triaza-spiro[4.5]decan-3-one;
2-(4-Difluoromethoxybenzyl)-l-(4-jfluorobenzyl)-8-(2-[l,3]dioxolan-2-yl- ethyl)- 1 ,2,8-triaza-spiro[4.5]decan-3-one;
8-Ethyl-l-(4-fluorobenzyl)-2-(4-isobutoxybenzyl)-l,2,8-triaza- spiro[4.5]decan-3-one; l-(4-Fluoroben2yl)-2-(4-isobutoxybenzyl)-8-isopropyl-l,2,8-triaza- spiro[4.5]decan-3-one;
8-Cyclopropylmethyl- 1 -(4-fluorobenzyl)-2-(4-isobutoxybenzyl)- 1 ,2,8- triaza-spiro[4.5]decan-3-one;
8-(2-[l,3]dioxolan-2-yl-ethyl)-l-(4-fluorobenzyl)-2-(4-isobutoxyben2yl)- l,2,8-triaza-spiro[4.5]decan-3-one;
4-(4-Ethoxybenzyl)-5-(4-fluorobenzyl)-9-methyl- 1 -oxa-4,9-diaza- spiro[5.5]undecan-3-one;
5-(4-Fluorobenzyl)-9-methyl-4-(4-propoxybenzyl)- 1 -oxa-4,9-diaza- spiro[5.5]undecan-3-one;
5-(4-fluorobenzyl)-4-(4-isobutoxybenzyl)-9-methyl- 1 -oxa-4,9-diaza- spiro[5.5]undecan-3-one;
5-(4-fluorobenzyl)-9-methyl-4-(4-trifluoromethoxybenzyl)- 1 -oxa-4,9- diaza-spiro[5.5]undecan-3-one;
5-(4-Chlorobenzyl)-4-(4-isobutoxybenzyl)-9-methyl-l-oxa-4,9-diaza- spirό[5.5]undecan-3-one;
5-(4-Chlorobenzyl)-4-(4-cyclopropylmethoxybenzyl)-9-methyl-l-oxa-4,9- diaza-spiro[5.5]undecan-3-one;
9-Ethyl-5-(4-fluorobenzyl)-4-(4-propoxybenzyl)- 1 -oxa-4,9-diaza- spiro [5.5]undecan-3 -one;
1 -(4-Fluorobenzyl)-2-(4-ethoxybenzyl)-9-methyl- 1 ,2,9-triaza- spiro [5.5]undecan-3 -one;
1 -(4-Fluorobenzyl)-2-(4-cyclopropylmethoxybenzyl)-9-methyl-l ,2,9- triaza-spiro[5.5]undecan-3-one;
1 -(4-Fluorobenzyl)-2-(4-isobutoxybenzyl)-9-methyl-l ,2,9-triaza- spiro[5.5]undecan-3-one;
1 -(4-Fluorobenzyl)-2-(4-propoxybenzyl)-9-methyl- 1 ,2,9-triaza- spiro[5.5]undecan-3-one; l-(4-Ethylbenzyl)-2-(4-isobutoxybenzyl)-9-methyl- l,2,9-triaza-spiro[5.5]undecan-3-one;
1 -(4-Fluorobenzyl)-2-(4-cyclopropylmethoxybenzyl)-9-ethyl- 1 ,2,9-triaza- spiro[5.5]undecan-3-one;
2-(4-Ethoxybenzyl)-l -(4-fluorobenzyl)-8-methyl-l ,2,4,8-tetraaza- sρiro[4.5]decan-3-one; 1 -(4-Fluorobenzyl)-2-(4-isobutoxyben2yl)-8-methyl- 1 ,2,4,8-tetraaza- sρiro[4.5]decan-3-one;
2-(4-Difluoromethoxybenzyl)- 1 -(4-fluorobenzyl)-8-methyl-2,8-diaza- spiro[4.5]decan-3-one; l-(4-Fluorobenzyl)-2-(4-isobutoxybenzyl)-8-methyl-2,8-diaza- spiro[4.5]decan-3-one ;
2-(4-Cyclopropylmethoxybenzyl)-l-(4-fluoroben2yl)-8-methyl-2,8-diaza- spiro [4.5] decan-3 -one;
8-Ethyl- 1 -(4-fluorobenzyl)-2-(4-isobutoxybenzyl)-2,8-diaza- spiro[4.5]decan-3-one;
8-(2-[l,3]Dioxolan-2-yl-ethyl)-l-(4-fluorobenzyl)-2-(4-isobutoxybenzyl)- 2,8-diaza-spiro[4.5]decan-3-one;
3 -(4-Difluoromethoxybenzyl)-4-(4-fluorobenzyl)- 8-methy 1- 1,3,8 -triaza- spiro [4.5]decan-2-one;
4-(4-Fluorobenzyl)-3-(4-isobutoxybenzyl)-8-methyl- 1 ,3,8-triaza- spiro [4.5] decan-2-one;
3-(4-Cyclopropylmethoxybenzyl)-4-(4-fluorobenzyl)-8-methyl-l,358- triaza-spiro [4.5] decan-2-one;
8-Ethyl-4-(4-fluorobenzyl)- 3-(4-isobutoxybenzyl)- 1 ,3 ,8-triaza- spiro[4.5]decan-2-one;
8-(2-[l,3]Dioxolan-2-yl-ethyl)-4-(4-fluorobenzyl)-3-(4-isobutoxybenzyl)- l,3,8-triaza-spiro[4.5]decan-2-one; l-(4-Fluorobenzyl)-2-(4-ethoxybenzyl)-9-methyl-2,4,9-triaza- spiro[5.5]undecan-3-one; l-(4-Fluorobenzyl)-2-(4-cyclopropylmethoxybenzyl)-9-methyl-2,4,9- triaza-spiro[5.5]undecan-3-one; l-(4-Fluorobenzyl)-2-(4-isobutoxybenzyl)-9-methyl-254,9-triaza- sρiro[5.5]undecan-3-one; l-(4-Fluorobenzyl)-2-(4-trifluoromethoxybenzyl)-9-methyl-2>4,9-triaza- spiro[5.5]undecan-3-one; l-(4-Fluorobenzyl)-2-(4-isobutoxybenzyl)-9-ethyl-2,4,9-triaza- spiro[5.5]undecan-3-one; 4-(4-Ethoxybenzyl)-5-(4-fluorobenzyl)-9-methyl-l-oxa-3,9-diaza- spiro[5.5]undecan-2-one;
3-(4-Ethoxybenzyl)-5-(4-fluorobenzyl)-9-methyl- 1 -oxa-3, 9-diaza- spiro[5.5]undecan-2-one;
3-(4-Ethoxybeiizyl)-4-(4-fluorobenzyl)-9-methyl- 1 -oxa-3 ,9-diaza- spiro[5.5]undecan-2-one;
5-(4-Fluorobenzyl)-4-(4-propoxybenzyl)-9-m ethyl- 1 -oxa-3 ,9-diaza- spiro [5.5]undecan-2-one;
5-(4-Fluorobenzyl)-3-(4-propxybenzyl)-9-methyl-l-oxa-3,9-diaza- spiro[5.5]undecan-2-one;
4-(4-Fluorobenzyl)-3 -(4-propoxybenzyl)-9-methyl- 1 -oxa-3 ,9-diaza- spiro[5.5]undecan-2-one;
5-(4-Fluorobenzyl)-4-(4-isobutoxybenzyl)-9-methyl- 1 -oxa-3,9-diaza- spiro[5.5]undecan-2-one;
4-(4-Fluoroben2yl)-3-(4-isobutoxybenzyl)-9-methyl- 1 -oxa-3, 9-diaza- spiro[5.5]undecan-2-one;
4-(4-Eth.oxybenzyl)-5-(4-fluorobenzyl)-9-methyl-2-oxa-4,9-diaza- spiro[5 -5]undecan-3 -one;
5-(4-Fluorobenzyl)-4-(4-methoxybenzyl)-9-methyl-2-oxa-4,9-diaza- spiro [5.5]undecan-3 -one;
5-(4-Fluoroben2yl)-4-(4-propoxyben2yl)-9-methyl-2-oxa-4,9-diaza- spiro [5.5]undecan-3 -one;
5-(4-Fluorobenzyl)-4-(4-isobutoxybenzyl)-9-methyl-2-oxa-4,9-diaza- spiro[5.5]undecan-3 -one;
4-(4-Ethoxybenzyl)-5-(4-fluorobenzyl)-9-methyl-2-oxa-4,9-diaza- spiro [5.5]undecan-3-one;
3-(4-Ethoxyben2yl)-4-(4-fluorobenzyl)-8-methyl-l-thia-3,8-diaza- spiro[4.5]decan-2-one;
4-(4-Fluorobenzyl)-3-(4-methoxybenzyl)-8-methyl-l-thia-338-diaza- spiro [4.5]decan-2-one;
4-(4-Fluorobenzyl)-3-(4-propoxybenzyl)-8-methyl-l-thia-3,8-diaza- spiro[4.5]decan-2-one; and 4-(4-Fluoroben2yl)-3-(4-isobutoxybenzyl)-8-methyl-l-thia-3,8-diaza- spiro[4.5]decan-2-one.
[0120] Other embodiments include use of compounds selected from the group consisting of:
2-(4-methoxyphenyl)-N-(4-methylbenzyl)-iV-{ 1 -[3-(1 ,3 dihydro-2H- benzimidazol-2-on-l-yl) propyl] piperidin-4-yl} acetamide;
1 -(4-methoxyphenyl)-N-(4-methylbenzyl)-N-(l -methylpiperidin-4- yl)cyclopropane carboxamide;
2-(4-Methoxyphenyl)-(l -phenylethyl)-N-(l -methylpiperidin-4-yl) acetamide;
2-(4-Methoxyphenyl)-N-(l-indanyl)-N-(l-methylpiperidin-4-yl) acetamide; and
2-(3,4-Methylenedioxyphenyl)-N-(4-methylbenzyl)-N-(l-methylpiperidin- 4-yl) acetamide.
[0121] The compounds of Formula I, II, and III exhibit activity at monoamine receptors, specifically serotonin receptors. Certain compounds share the common property of acting as inverse agonists at the 5-HT2A receptor. Thus, experiments performed on cells transiently expressing the human phenotype of the receptor have shown that the compounds of general Formula I, II, and III attenuate the signaling of such receptors in the absence of additional ligands acting upon the receptor. The compounds have thus been found to possess intrinsic activity at this receptor and are able to attenuate the basal, non- agonist-stimulated, constitutive signaling responses that the 5-HT2A receptor displays. The observation that the compounds of general Formula I, II, and III are inverse agonists also indicates that these compounds have the ability to antagonize the activation of 5- HT2A receptors that is mediated by endogenous agonists or exogenous synthetic agonist ligands.
[0122] Therefore, some embodiments include use of compounds of Formula I, II, III and the salts and stereoisomers thereof, including compounds that show a relatively high degree of selectivity towards the 5-HT2A subtype of serotonin receptors relative to other subtypes of the serotonin (5-HT) family of receptors as well as to other receptors, most particularly the monoaminergic G-protein coupled receptors, such as dopamine receptors. In one embodiment, these compounds act as inverse agonists and/or antagonists at the 5-HT2A subtype of serotonin receptors. Methods of preparation
[0123] The compounds of Formulae I and II may in general be prepared by routes such as those summarized below, or by modification of these methods. Ways of modifying the methodology include, among others, temperature, solvent, reagents etc, and will be obvious to those skilled in the art.
[0124] For instance, compounds of the Formula C may be synthesized from the corresponding ketone A by reductive animation utilizing any primary amine. The reaction is conveniently carried out by stirring the reactants in an inert solvent such as methanol or ethanol containing acetic acid. As reducing agent NaBH4, NaCNBHb, BH3-pyridine or any related reagent may be used including solid-supported reagents. The reaction is typically carried out at room temperature. The ketone A, as exemplified by the piperidone, may be chosen from a list of compounds corresponding to the Z and Zi- groups listed in Formulae I and II. The ketones can either be obtained commercially or synthesized by methodology disclosed in Lowe et al. J. Med. Chem. 37: 2831-40 (1994); Carroll et al. J. Med. Chem. 35:2184-91 (1992); or Rubiralta et al. Piperidine - Structure, Preparation, Reactivity and Synthetic Applications of Piperidine and its Derivatives. (Studies in Organic Chemistry 43, Elsevier, Amsterdam, 1991). The protecting group P includes groups such as those described in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Chemistry, 3. Ed. John Wiley & Sons, 1999, and they should be chosen in such a way, that they are stable to the reaction conditions applied and readily removed at a convenient stage using methodology known from the art. Typical protecting groups are N-Boc, N-Cbz, N-Bn.
[0125] Alternatively, the amine C can be synthesized from the primary amine B by reductive amination with any aldehyde. The reaction is conveniently carried out by stirring the reactants in an inert solvent such as methanol or ethanol containing acetic acid. As reducing agent NaBH4, NaCNBHa, BH3-pyridine or any related reagent may be used including solid-supported reagents. The reaction is typically carried out at room temperature. The primary amine B, as exemplified by the 4-aminopiperidine, may be chosen from a list of compounds corresponding to the Z and Zi-groups listed in Formulae I and II. The amines can either be obtained commercially or synthesized from the corresponding ketones. The protecting group P may be chosen as stated above. [0126] Alternatively, the amine C can be synthesized from the primary amine B by alkylation with any alkylating agent (R-L1). The leaving group Li is suitably a halogen atom, e.g., bromine or iodine, or a sulfonate, e.g. tosylate or mesylate, or another leaving group favoring the reaction. The reaction is conveniently carried out by stirring the reagents under basic conditions in an inert solvent, e.g., diisoprόpylethylamine in acetonitrile, or K2CO3 in ΛζN-dimethylformamide. The reaction is typically carried out at temperatures between room temperature and 8O0C. The primary amine B, as exemplified by the 4-aminopiperidine, may be chosen from a list of compounds corresponding to the Z and Zl -groups listed in Formulae I and II. The amines can either be obtained commercially or synthesized from the corresponding ketones. The protecting group P may be chosen as stated above.
Figure imgf000055_0001
Figure imgf000055_0002
Figure imgf000055_0003
[0127] Wherein R and R* are defined in agreement with Formulae I and II, and P represents a suitable protecting group, and Li represents a suitable leaving group.
[0128] The secondary amine C may be acylated using any isocyanate or isothiocyanate (QpN=C=W or Qi-N=C=Wi) to give the corresponding ureas or thioureas D. The reaction is typically carried out by stirring the reactants, using an excess of isocyanate or isothiocyanate in an inert solvent, e.g., dichloromethane at a temperature between 00C and room temperature and under dry conditions. The amine C may also be acylated using any carboxylic acid halide (Q2COX), e.g., chloride, or carboxylic anhydride ((Q2C=O)2O) to give amides of the general structure E. The reaction is typically carried out using an excess of the acylating agent and a suitable base, e.g., triethylamine or diisopropylethylamine in an inert solvent, e.g., dichloromethane, at a temperature between O0C and room temperature and under dry conditions. As an alternative to the carboxylic acid halides and carboxylic acid anhydrides, the amine C may be acylated using a carboxylic acid (Q2COOH) and a suitable coupling reagent e.g. PyBroP, DCC or EDCI. The reaction is typically carried out using an excess of the acylating agent and the coupling reagent in an inert solvent, e.g., dichloromethane at a temperature between O0C and room temperature and under dry conditions. The compounds of the general structure (E) may be converted into the corresponding thioamides using methodology disclosed in Varma et al., Org. Lett. 1: 697-700 (1999); Cherkasov et al. Tetrahedron 41:2567 (1985); or Scheibye et al, Bull. Soc. Chim. BeIg. 87:229 (1978).
Figure imgf000056_0001
Figure imgf000056_0002
[0129] Wherein R, Qi, Q2, W and Wi are defined in agreement with Formulae I and II, P represents a suitable protecting group, and X represents a halide.
[0130] The substituent T on the ring nitrogen in compounds F or G can be introduced by a two step procedure. First, the protecting group on the urea D or the amide E is removed using well-known methods. For example, the N-Boc group is removed by treating the protected compound with 4 M HCl in dioxane or trifluoroacetic acid in dichloromethane. Second, the secondary amines obtained from D and E can be alkylated by reductive amination using any aldehyde (T*-CHO) or ketone (T=O). The reaction is conveniently carried out by stirring the reactants in an inert solvent such as methanol or ethanol. As a reducing agent, solid-supported borohydride, NaBH4, NaCNBH3, BH3 -pyridine, H2/Pd-C or any related reagent may be used, including solid-supported reagents. The reaction is typically carried out at room temperature."
[0131] Alternatively, the compounds F and G can be synthesized from the secondary amine obtained from D or E as described above by alkylation with any alkylating agent (T- Lj). The leaving group Li is suitably a halogen atom, e.g., bromine or iodine, or a sulfonate, e.g., tosylate or mesylate, or another leaving group favoring the reaction. The reaction is conveniently carried out by stirring the reagents under basic conditions in an inert solvent, for example diisopropylethylamine in acetonitrile, or K.2CO3 in NjN-dimethylforrnamide. The reaction is typically carried out at temperatures between room temperature and 8O0C.
[0132] Alternatively, the T-group can be introduced in the first step of the synthetic sequence leading to the compounds in accordance with the present invention by JV-alkylation of compound H with any alkylating agent (T- Li). The leaving group L1 is suitably a halogen atom, e.g., bromine or iodine, or a sulfonate, e.g., tosylate or mesylate, or another leaving group favoring the reaction. The reaction is conveniently carried out by stirring the reagent under basic conditions in an inert solvent, e.g., diisopropylethylamine in acetonitrile, or K23 in N,N-dimethylformamide. The reaction is typically carried out at temperatures between room temperature and 8O0C. Alternatively the T-group can be introduced in the first step by reductive amination using any aldehyde (T* -CHO) or ketone (T=O) and a suitably protected compound H', exemplified by 4-piperidone ethylene ketal. The reaction is conveniently carried out by stirring the reactants in an inert solvent such as methanol or ethanol. As a reducing agent, solid-supported borohydride, NaBH4, NaCNBH3, BH3-pyridine, H2ZPd-C or any related reagent may be used, including solid-supported reagents. The reaction is typically carried out at room temperature, but less reactive carbonyl compounds may require higher temperatures and/or the preformation of the corresponding imine under water removal before addition of the reducing agent. Removal of the protecting group gives the desired compound J. The secondary amine H and H1, as exemplified by 4-piperidone and its protected derivative, may be chosen from a list of compounds corresponding to the Z and Zl -groups listed in Formulae I and II. The amines can either be obtained commercially or synthesized from methodology disclosed in Lowe et al., J. Med. Chem. 37:2831-40 (1994); and Carroll et al., J. Med. Chem. 35:2184-91 (1992).
[0133] Alternatively, compounds of the general structure J may be synthesized starting from K using the method disclosed in: Kuehne et al., J. Org. Chem. 56:2701 (1991); and Kuehne et al., J. Org. Chem. (1991), 56:513.
Figure imgf000058_0001
Figure imgf000058_0002
Figure imgf000058_0003
Figure imgf000058_0004
[0134] Wherein R, Q1 , Q2, W, and T are defined in agreement with Formulae I and II, and Li is a suitable leaving group.
[0135] Heterocyclylalkyl alkylating agents such as T-Li may be commercially available or are typically obtained by alkylation of a heterocycle with a bifunctional alkyl- linker, as shown below. The leaving groups Li and L2 are suitably a halogen atom, e.g., chlorine, bromine or iodine, or a sulfonate, e.g., tosylate or mesylate, or another leaving group favoring the reaction. The reaction is conveniently carried out by stirring the reagent under basic conditions in an inert solvent, e.g., diisopropylethylamine in acetonitrile, or K2CO3 in NvN-dimethylformamide. The reaction is typically carried out at temperatures between room temperature and 8O0C. The alkylating agent hence obtained can be either reacted in situ in the next step with the secondary amine (i.e. deprotected D/E, or H) or isolated from the reaction mixture before its further use. Heterocyclylalkyl alcohols such as T+-CH2OH or T-OH may also be converted into suitable alkylating agents T-Li by transforming the hydroxyl into a leaving group, e.g. by tosylation, mesylation or halogenation. Alternatively, T+-CH2OH or T-OH may be oxidized to the corresponding aldehydes or ketones T+-CHO or T=O with, for example, pyridinium chlorochromate, CrOa-H2SO4, or via the Swera or Dess-Martin procedures, to be used in a reductive animation step with the secondary amines as described above.
Figure imgf000059_0001
[0136] Wherein Y5 p and T are defined in agreement with Formulae I and II, and Li and L2 are suitable leaving groups.
[0137] The building blocks incorporating the aromatic groups Ari, Ar2, Ar3 and
4 may either be obtained commercially or synthesized from methodology disclosed in the literature. The introduction of substituents on Ari, Ar2> Ar3 and/or Ar4 may be performed from a suitable precursor at any appropriate stage of the preparation of the compounds.
[0138] For instance, compounds containing an alkoxy substituents may be typically prepared by Williamson ether synthesis from the corresponding hydroxyaryl derivatives. [0139] Structures bearing an amine substituent on Ari, Ar2, Ar3 and/or Ar4 may be obtained from a suitable halo- or pseudohalo precursor (e.g. Br, I-, Cl-, triflate-, nonaflate-, tosylate-substituted aryl derivatives) by metal-catalyzed amination chemistries, such as Pd- or Ni- (Hartwig, Angew. Chem. Int. Ed., 1998, 37, 2046-2067; Yang & Buchwald, J. Organometallic Chem., 1999, 576, 125-146; Hartwig in Modern Amination Methods; Ricci, Ed.; Wiley- VCH: Weinheim, Germany, 2000) or Cu-catalyzed (Buchwald et al, Org. Lett., 2002, 4, 5% 1-584; Kwong & Buchwald, Org. Lett., 2003, 5, 793-796). Alternatively, these compounds can be obtained from aniline-based precursors either by alkylation (Hickinbottom, J. Chem. Soc. 1930, 992), or by reductive amination (Emerson & Walters, J. Am. Chem. Soc, 1938, 60, 2023; Milovic et al, Synthesis, 1991, 11, 1043-1045), or by dehydrative alkylation (Rice & Kohn, J. Am. Chem. Soc, 1955, 77, 4052; Brown & Reid, J. Am. Chem. Soc, 1924, 46, 1838). Additionally, compounds of this type may also be synthesized from corresponding boronic acids by Cu-catalyzed coupling (Antilla & Buchwald, Org. Lett., 2001, 3, 2077-2079).
[0140] The structures bearing an amide substituent on Arj, Ar2, Ar3 and/or Ar4 may be obtained from a suitable halo- or pseudohalo precursor either by Pd catalyzed (Yin & Buchwald, J. Am. Chem. Soc, 2002, 124, 6043-6048) or by Cu catalyzed (Buchwald et al, J. Am. Chem. Soc, 2002, 124, 7421-7428) amidation chemistries. Alternatively, these compounds may also be obtained from the corresponding aniline precursors either by acylation (Wolf, Liebigs Ann. Chem., 1952, 576, 35; Yasukara et al, J. Chem. Soc. Perkin Trans. 1, 2000, 17, 2901-2902; Nigam & Weedon, J. Chem. Soc, 1957, 2000) or by formylation (Hirst & Cohen, J. Chem. Soc, 1895, 67, 830; Olah & Kuhn, Chem. Ber. 1956, 89, 2211; Guthrie et al, Can. J. Chem., 1993, 71, 2109-2122).
[0141] Compounds that carry an alkylsulfanyl substituent on Arj, Ar^ Ar3 and/or Ar4 be obtained from a suitable halo- or pseudohalo precursor by Pd catalyzed (Li, J. Org. Chem., 2002, 67, 3643-3650), or Cu catalyzed (Kwong & Buchwald, Org. Lett., 2002, 4, 3517-3520) thioetherification chemistry. Alternatively, these compounds may be prepared by alkylation of corresponding benzenethiol precursors (Vogel, J. Chem. Soc, 1948, 1809; Landini & Rocca, Synthesis, 1974, 565-566; Bun-Hoi et al, J. Org. Chem., 1951, 16, 988). Alternatively, alkylarylsulfanyls may be obtained by irradiation of benzenethiols and alkenes (Screttas & Micha-Screttas, J. Org. Chem., 1978, 43, 1064- 1071). [0142] Compounds that bear an acyl group on An, Ar2, A^ and/or Ar4 may be prepared from corresponding aryl iodides by Pd catalyzed (Cacchi et al, Org. Lett, 2003, 5, 289-293) acylation chemistry. Alternatively, they may be obtained from the corresponding benzenes by Friedel-Crafts chemistry (Read, J. Am. Chem. Soc, 1922, 44, 1746-1755), or by addition of aryl-Grignard reagents to nitriles (Whitmore et al, J. Am. Chem. Soc, 1947, 69, 235-237) or to acyl chlorides (Whitmore & Lester, J. Am. Chem. Soc, 1942, 64, 1247), or by either Pd-catalyzed (Gooβen & Ghosh, Angew. Chem. Int. Ed. Engl., 2001, 40, 3458-3460) or Rh-catalyzed acylation of arylboronic acids.
[0143] Compounds that bear an iV-containing aromatic heterocycle on Ari, Ar2j Ar3 and/or Ar4 can be obtained either by metal-catalyzed cross-couplings (Buchwald et al, Org. Lett., 2002, 2, 1403-1406; Buchwald et al, J. Am. Chem. Soc, 2001, 123, 7727 '-7729; Buchwald et al, J. Am. Chem. Soc, 2002, 124, 11684-11688). Alternatively, they may be accessed from suitable precursors such as aryl hydrazines, aryl amines or aryl nitriles according to literature procedures (e.g. Alvisi, Gazz. Chem. Ital., 1892, 22, 159; Finar, Godfrey, J. Chem. Soc, 1954, 2293; Muri et al. Synth. Commun., 1998, 28, 1299- 1321; Artico et al, Europ. J. Med. Chem. Chim. Ther., 1992, 27, 219-228; Biagi et al., Farmaco Ed. Sci. 1988, 43, 597-612; Stefancich et al, Farmaco Ed. Sci., 1984, 39, 752- 764).
[0144] The compounds of Formula III may in general be prepared by routes such as those summarized below, or by modification of these methods. Cyclization of the appropriate intermediates may be generally achieved with phosgene or its analogues such as CDI, with chloroacetylchloride or equivalents thereof or by treatment with carbondisulfϊde and subsequent oxidation.
[0145] Introduction of the desired piperidine substituent (R9) can generally be achieved, after N-deprotection if required, by alkylation or by reductive amination.
[0146] Compounds of Formula III in which m" = 1, Z3 = CH and X4 = O or OCH2 may be synthesized from suitably protected 4-piperidone as described by Bassus et al. (Eur. J. Med. Chem. - Chim. Ther. 9:416-423 (1974)). Alkylation of the carbamate nitrogen and introduction of the desired piperidine iV-substituent leads to the 3,5- disub.stituted spirocycle. 3,4 or 4,5-disubstituted derivatives may be prepared as described by Fisera et al. (Monatsh. Chem. 125:909-919 (1994)), via an appropriately substituted isoxazoline, followed by reductive cleavage. Cyclisation of the resulting γ-aminoalcohols gives the desired spirocycles. Alternatively, 4,5-disubstituted or 3,4,5-trisubstituted spirocycles may be prepared from an appropriate β-ketoester by reaction with a halide to introduce the 5-substituent, reductive amination (with a primary amine for the 3- substituent), treatment with allyl magnesium bromide, cyclisation as described above, oxidative cleavage of the double bonds (e.g. by ozonolysis) and formation of the piperidine ring by reductive amination.
[0147] Compounds of Formula III in which m" = 1, Z3 = N and X4 = O or OCH2 may be prepared by a Strecker synthesis involving an appropriate aldehyde and e.g. tert-bυtyl carbazate. The nitrile may be transformed into an ester, which is reacted with allyl magnesium bromide, followed by oxidative cleavage of the two olefins, formation of the piperidine by reductive amination. Alkylation, deprotection of the hydrazide and cyclisation gives the desired spirocycles. Depending on the desired substituents and their positions, these steps may be inversed and additional protection steps of functional groups may be required. Alternatively, the compounds may be obtained from an appropriate β- ketoester by reaction with a bis(2-chloroethyl) amine derivative to form the piperidine ring. Reductive amination, saponification and Curtius rearrangement lead to the cyclic urea derivative.
[0148] Compounds of Formula ffl in which m" = I3 Z3 = N and X4 = N(R) may be prepared adapting methods described by Bhatia et al. (J. Med. Chem. 39:3938- 3950 (1996)) starting from the suitably protected Strecker-product 4-amino-4-cyano- piperidine.
[0149] Compounds of Formula III in which m" = 0, Z3 = CH and X4 = CH2CH2 may be synthesized by Michael addition of a nitrile-derivative to suitably protected 4-methoxycarbonyl-methylenepiperidine, reduction of the nitrile-group to the amine, followed by lactam ring formation, alkylation of the resulting amide and introduction of the desired piperidine substituent.
[0150] Compounds of Formula III in which m" = 0, Z3 = CH and X4 = CH2O may be prepared from the appropriate β-ketoester by reaction with a suitably protected bis(2-chloroethyl) amine. Reductive amination, followed by reduction of the nitrile to the alcohol followed by cyclisation and introduction of the piperidine substituent by alkylation or reductive amination leads to the desired spirocycle.
[0151] Compounds of Formula III in which m" = 0, Z3 = CH and X4 = CH2N may be prepared from the appropriate β-ketonitrile by reaction with a suitably protected bis(2-chloroethyl) amine. Reductive animation, followed by reduction of the nitrile to the amine followed by cyclisation and introduction of the piperidine substituent by alkylation or reductive animation leads to the desired spirocycle.
[01521 Compounds of Formula III in which m" = 0, Z3 = CH and X4 = OCH2 may be prepared as described in herein.
[01531 Compounds of Formula III in which m" = 0, Z3 = N and X4 = CH2CH2 may be prepared from protected 8-aza-l-oxa-spiro[4.5]decan-2-one (Mullen et al. J. Med. Chem. 43:4045 (2000)) by reaction with hydrazine, then S-ethyltrifluorothioacetate, followed by a Mitsunobu reaction according to Meng et al. (Tetrahedron 47:6251 (1991)). The appropriate substituents in 1,2- and 9-positions are introduced by alkylation and/or reductive amination.
[0154] Compounds of Formula III in which m" = 0, Z3 = N and X4 = CH2O may be prepared by a Strecker synthesis from suitably protected 4-piperidone and a carbazate (or an appropriate hydrazine). Transformation of the resulting nitrile into the alcohol, cyclisation using methods described above, alkylation (after basic hydrolysis of the exocyclic carbazate function if present) and introduction of the piperidine N- substituent leads to the desired compound.
[01551 Compounds of Formula III in which m" = 0, Z3 = N and X4 = CH2N may be prepared using the same strategy as for the preparation of 1 ,2,9-triaza-4-oxa- spiro[5.5]undecan-3-one, except that the nitrile is reduced to the corresponding amine.
[0156] Compounds of Formula III in which m" = 0, Z3 = CH and X4 = CH2 may be synthesized by Michael addition of a nitro-derivative to suitably protected 4- methoxycarbonyl-methylenepiperidine, reduction of the nitro-group to the amine, followed by lactam ring formation, alkylation of the resulting amide and introduction of the desired piperidine substituent.
[0157] Compounds of Formula III in which m" = 0, Z3 = CH and X4 = O may be prepared as described in herein. Alternatively, a nitroaldol reaction may be used to obtain the desired intermediate 1,2-aminoalcohol after reduction of the nitro-group, followed by cyclisation. Alternatively, the compounds may be prepared by epoxidation of an appropriate olefin, obtained by Wittig or Homer- Wads worth-Emmons reactions from suitably protected 4-piperidone. Epoxide opening with ammonia or a primary amine, followed by cyclisation with a phosgene equivalent, alkylation of the carbamates if required and introduction of the desired piperidine substituent by alkylation or reductive animation leads to the target compound. Besides the preparation of enantiopure compound as described in herein, an enantioselective modification may include an asymmetric epoxidation method of the olefin as described in the literature, e.g. Jacobsen or others. Alternatively, Sharpless asymmetric dihydroxylation method may be used followed by epoxide ring formation. Alternatively, suitably protected 4-methoxycarbonyl- methylenepiperidine may be reduced to the allyl alcohol which is subjected to Sharpless asymmetric epoxidation according to literature procedure. Epoxide opening with a metallorganic reagent and oxidation of the resulting primary alcohol leads to the β- hydroxy carboxylic acid, which is converted into the desired spirocyclic enantiomer as described in herein. Alternatively, suitably protected 4-methoxycarbonyl- methylenepiperidine may be converted to an enantiomerically pure epoxide by Jacobsen epoxidation, followed by ring opening with ammonia or an appropriate amine, reaction with a metallorganic reagent with the ester group, reduction, cyclisation, alkylation if required and introduction of the piperidine substituent. Alternatively, the stereocenter may be introduced by using an appropriate α-amino acid ester as the chiral template. Reaction with allyl magnesium bromide, cyclisation, oxidative cleavage of the allyl groups, formation of the piperidine ring by reductive amination and final alkylation of the carbamate leads to the desired enantiomerically pure derivative.
[0158] Compounds of Formula III in which m" = 0, Z3 = CH and X4 = N or NCEb may be prepared by a Strecker synthesis involving suitably protected 4-piperidone and an appropriate primary amine. Reaction of the resulting nitrile with a Grignard reagent gives the ketone, which is then subjected to a reductive amination. Cyclisation of the resulting diamine (after deprotection step if necessary), with phosgene or an equivalent thereof leads to the cyclic urea. Alkylation or reductive amination steps may be used to introduce N-piperidine substituents. In a similar way, the 6-membered analogue (X4 = NCH2) may be obtained by using chloroacetyl chloride or a similar reagent for cyclisation (additional use of protecting groups may be necessary).
[0159] Compounds of Formula III in which m" = 0, Z3 = N and X4 = CH2 may be prepared as outlined herein.
[0160] Compounds of Formula III in which m" = 0, Z3 = N and X4 = N(R) may be obtained according to literature methods (Gstach et al. Synthesis 803-808 (1990)) by treatment of a suitably protected 4-ρiperidone by reaction with a hydrazine derivative and reaction of the intermediate hydrazone with potassium cyanate. Alkylation and/or reductive amiπation after deprotection may be used to introduce the desired substituents.
[0161] Alternatively, compounds of Formula III in which X4 = S or SCH2 or CH2S may be obtained as described by routes described for the equivalent compounds bearing X4 = O, OCH2 or CH2O by transforming, prior to cyclisation, the appropriate hydroxyl group into a thiol using well known literature procedures (e.g. treatment with acetyl chloride and substitution with benzylthiol and conversion to the free mercaptane). Similarly, the hydroxyl group may be converted into the corresponding amines, which constitutes an alternative way to access some compounds of Formula III in which X4 = N(R), N(R)CH2, CH2(R).
[0162] Compounds of Formula III in which W2 = S may be prepared from the corresponding compounds in which W2 = O by treatment with e.g. the Lawesson reagent or bis(tricyclohexyltin)sulfide and BCl3 or other sulfur-transferring reagents.
[0163] In general, during any of the processes for preparation of the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic Chemistry (ed. J.F.W. McOmie, Plenum Press, 1973); and T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Chemistry, 3. Ed. John Wiley & Sons, 1999, and they should be chosen in such a way, that they are stable to the reaction conditions applied and readily removed at a convenient stage using methodology known from the art. Typical protecting groups are N-Boc, N-Cbz, N-Bn.
Pharmaceutical Compositions
[0164] Other embodiments includes the use of pharmaceutical compositions comprising a compound as described above, and a physiologically acceptable carrier, diluent, or excipient, or a combination thereof.
[0165] The term "pharmaceutical composition" refers to a mixture of a compound disclosed herein with other chemical components, such as diluents or carriers. The pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to, oral, intramuscular, intraocular, intranasal, intravenous, injection, aerosol, parenteral, and topical administration. Pharmaceutical compositions can also be obtained by reacting compounds with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like. Pharmaceutical compositions will generally be tailored to the specific intended route of administration.
[0166] The term "physiologically acceptable" defines a carrier or diluent that does not abrogate the biological activity and properties of the compound.
[0167] The pharmaceutical compositions described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or suitable carriers or excipient(s). Techniques for formulation and administration of the compounds of the instant application may be found in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA3 18th edition, 1990, which is hereby incorporated by reference in its entirety.
[0168] Suitable routes of administration may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal, intraocular injections or as an aerosol inhalant.
[0169] Alternately, one may administer the compound in a local rather than systemic manner, for example, via injection of the compound directly into the area of pain or inflammation, often in a depot or sustained release formulation. Furthermore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with a tissue-specific antibody. The liposomes will be targeted to and taken up selectively by the organ.
[0170] The pharmaceutical compositions disclosed herein may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes.
[0171] Pharmaceutical compositions for use in accordance with the present disclosure thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations, which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., as disclosed in Remington's Pharmaceutical Sciences, cited above.
[0172] For injection, the agents disclosed herein may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
[0173] For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds disclosed herein to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by mixing one or more solid excipient with pharmaceutical combination disclosed herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
[0174] Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
[0175] Pharmaceutical preparations, which can be used orally, include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
[0176] For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.
[0177] For administration by inhalation, the compounds for use according to the present disclosure are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
[0178] The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
[0179] Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents, which increases the solubility of the compounds to allow for the preparation of highly, concentrated solutions.
[0180] Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
[0181] The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides. [0182] In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
[0183] A pharmaceutical carrier for the hydrophobic compounds disclosed herein is a co-solvent system comprising benzyl alcohol, a nonpolar surfactant, a water- miscible organic polymer, and an aqueous phase. A common co-solvent system used is the VPD co-solvent system, which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80™, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. Naturally, the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics. Furthermore, the identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of Polysorbate 80™; the fraction size of polyethylene glycol may be varied; and other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone. Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed.
[0184] Many of the compounds used in the pharmaceutical combinations disclosed herein may be provided as salts with pharmaceutically compatible counterions. Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free acids or base forms.
[0185] Pharmaceutical compositions suitable for use in the methods disclosed herein include compositions where the active ingredients are contained in an amount effective to achieve its intended purpose. More specifically, a "therapeutically effective amount" means an amount of compound effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
[0186] The exact formulation, route of administration and dosage for the pharmaceutical compositions disclosed herein can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl et al. 1975, in "The Pharmacological Basis of Therapeutics", Chapter 1, which is hereby incorporated by reference in its entirety). Typically, the dose range of the composition administered to the patient can be from about 0.5 to 1000 mg/kg of the patient's body weight, or 1 to 500 mg/kg, or 10 to 500 mg/kg, or 50 to 100 mg/kg of the patient's body weight. The dosage may be a single one or a series of two or more given in the course of one or more days, as is needed by the patient. Where no human dosage is established, a suitable human dosage can be inferred from ED50 or ID50 values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals.
[0187] Although the exact dosage will be determined on a drug-by-drug basis, in most cases, some generalizations regarding the dosage can be made. The daily dosage regimen for an adult human patient may be, for example, an oral dose of between 0.1 mg and 500 mg of each ingredient, preferably between 1 mg and 250 mg, e.g. 5 to 200 mg or an intravenous, subcutaneous, or intramuscular dose of each ingredient between 0.01 mg and 100 mg, preferably between 0.1 mg and 60 mg, e.g. 1 to 40 mg of each ingredient of the pharmaceutical compositions disclosed herein or a pharmaceutically acceptable salt thereof calculated as the free base, the composition being administered 1 to 4 times per day. Alternatively the compositions disclosed herein may be administered by continuous intravenous infusion, preferably at a dose of each ingredient up to 400 mg per day. Thus, the total daily dosage by oral administration of each ingredient will typically be in the range 1 to 2000 mg and the total daily dosage by parenteral administration will typically be in the range 0.1 to 400 mg. In some embodiments, the compounds will be administered for a period of continuous therapy, for example for a week or more, or for months or years.
[0188] Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety, which are sufficient to maintain the modulating effects, or minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
[0189] Dosage intervals can also be determined using MEC value. Compositions should be administered using a regimen, which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%.
[0190] In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.
[0191] The amount of composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
[0192] The compositions may, if desired, be presented in a pack or dispenser device, which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert. Compositions comprising a compound disclosed herein formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
EXAMPLES
[0193] The invention is disclosed in further detail in the following examples that are not in any way intended to limit the scope of the invention as claimed. Chemical Synthesis
[0194] General procedures. 1H NMR spectra were recorded at 400 MHz on a Varian Mercury- VX400MHz spectrometer. 13C NMR spectra were measured at 100 MHz with proton decoupling at ambient temperature. Chemical shifts are given in δ-values [ppm] referenced to the residual solvent peak chloroform (CDCI3) at 7.26 and methanol (CD3OD) at 3.31 ppm. Unless otherwise stated, the NMR spectra of the compounds are described for their free amine form. Due to the presence of rotamers, two sets of signals are generally observed and rotamer ratios are reported. Where the corresponding signals for each of the two rotamers could unmistakably be identified, they are reported together [e.g. 4.66-4.58 and 3.76-3.68 (2m, IH)].
[0195] Materials and solvents were of the highest grade available from commercial sources and used without further purification. Acidic ion-exchange solid phase extraction (SPE) cartridges were MEGA BE-SCX from Varian.
General LC-MS procedure for Examples 1-41 :
[0196] All spectra were obtained using an HPI lOO LC/MSD-instrument. A setup with a binary pump, autosampler, column oven, diode array detecter, and electrospray ionization interface was used. A reversed phase column (C 18 Luna 3 mm particle size, 7.5 cm X 4.6 mm ID) with a guard cartridge system was used. The column was maintained at a temperature of 300C. The mobile phase was acetonitrile/8 mM aqueous ammonium acetate. A 15 minute gradient program was used, starting at 70% acetonitrile, over 12 minutes to 95% acetonitrile, over 1 minute back to 70% acetonitrile, where it stayed for 2 minutes. The flow rate was 0.6 ml/min. The tr values reported in the specific examples below were obtained using this procedure.
Example 1 - N-((4-Methylphenyl)methyl)-N-rpiperidin-4-yl)-N'-phenylmethylcarbamide r26HCH65)
[0197] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (1.75 g, 8.8 mmol)and 4-methylbenzylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (1 M5 6.7 ml) followed by NaCNBH3 in methanol (0.3 M, 30 ml). The resulting solution was stirred at room temperature. After 20 h, water (5 ml) was added, and the mixture was stirred for 1 h before it was concentrated. Flash chromatography in dichloromethanermethanol 10:1 gave tert-butyl 4-(4- methylphenyl)methyl)amino-piρeridine carboxylate. Yield: 2.4 g, 98%. To a solution of tert-butyl 4-(4-methylphenyl)methyl)amino-piperidine carboxylate (800 mg, 2.63 mmol) in dry dichloromethane (20 ml) was added benzylisocyanate (0.65 ml, 5.26 mmol). The solution was stirred at room temperature. After 48 h, an excess of 2- dimethylaminoethylamine was added. The mixture was stirred for another 24 h, before it was concentrated. The resulting solid was redissolved in dichloromethane (20 ml), sequentially washed with HCl (0.2 N5 3 X 30 ml), and water (20 ml), dried (Na2SO4), filtered and concentrated. Flash chromatography in dichloromethanermethanol 10:1 gave N-((4-methylphenyl)methyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-N'- phenylmethylcarbamide (760 mg, 66%), which was dissolved in diethyl ether (5 ml). HCl (4 M) in dioxane (3 ml) was added, and the solution was stirred at room temperature for 60 min, then concentrated. The resulting oil was redissolved in a mixture of dichloromethane and diethyl ether (4:1). The organic layer was extracted with HCl (0.2 M, 3 X 20 ml). The combined aqueous layers were treated with NaOH (0.2 M) until basic (pH>8), then extracted with dichloromethane (3 X 20 ml). The combined organic layers were dried (Na2SO4), filtered, and concentrated to give the title compound. Yield: 406 mg, 70%; 13C-NMR (CDCl3): δ 21.3, 31.6, 45.0, 45.9, 46.4, 53.0, 126.3, 127.2, 127.4, 128.6, 129.8, 135.3, 137.4, 139.7, 158.5.
Example 2 - N-('(4-Methylphenyl)methyl)-N-(l-('2-methylpropyl)piperidin-4-yl)-N'- phenylme thylcarbamide f26HCH66-02)
[0198] The product from example 1 above (20 mg, 0.06 mmol)was dissolved in abs. ethanol (2 ml). 2-Methylpropionaldehyde (0.08 ml, 0.6 mmol)was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol)was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. IR: 1640, 1 185, 1110 cm'1; LC-MS: (M+H)+ 394.2, tr 5.60 min. Example 3 N-(I -((2-BromoDhenyl>methyl')piperidin-4-yl')-N-('('4-methylphenyl')methyl)- N'-p henylmethylcarbamide f26HCH66-03~)
[01991 The product from example 1 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml). 2-Bromobenzaldehyde (0.07 ml, 0.6 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0:3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. IR: 1635, 1180, 1110 cm"1; LC-MS: (MH-H)+ 506.1, tr 8.37 min.
Example 4 - N-(l-('(4-Hvdroxy-3-methoxyphenyl)methyπpiperidin-4-yl)-N-r(4- methylphenvD methyl VN '-phenylmethylcarbamide (26HCH66-04)
[0200] The product from example 1 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml). 4-Hydroxy-3-methoxybenzaldehyde (91 mg, 0.6 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. 13C-NMR (CD3OD, selected): δ 19.9, 55.4, 126.5, 127.0, 128.1, 129.0, 140.3, 148.0, 148.1, 158.8.
Example 5 : N-C 1 -f (5-Emylthien-2-yl)memvnpiperidin-4-yiyN-(T4- methylphenyl')methvn- N'-phenylmethylcarbamide (26HCH66-05)
[0201] The product from example 1 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml). 5-Ethyl-2-thiophencarboxaldehyde (84 mg, 0.6 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. IR: 1640, 1185, 1110, 805, 700, 620 cm"1; LC- MS: (M+H)+ 462.3, tr 7.52 min.
Example 6 - N-(l-(Imidazol-2-ylmethyl)piperidin-4-vπ-N-r(4-methylphenyl)methvπ-N'- phe nylmethylcarbamide C26HCH66-06)
[0202] The product from example 1 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml). Imidazole-2-carboxaldehyde (58 mg, 0.6 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. IR: 1620, 1190, 1100, 805, 700, 620 cm"1; LC- MS: (M+H)+ 418.2, tr 2.05 min.
Example 7 - N-(l-(Cyclohexylmethyl')piperidin-4-yl)-N-((4-methylphenyl)methyl)-N'- phenyl methylcarbamide f26HCH66-09)
[0203] The product from example 1 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml). Cyclohexanecarboxaldehyde (67 mg, 0.6 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. IR: 1635, 1175, 1 100, 805, 695, 620 cm"1; LC- MS: (M+H)+ 434.4, tr 7.44 min. Example 8 - N-π-r(4-Fluorophenyl)methvπpiperidin-4-ylVN-('('4-methylphenvπmethyπ- N'- phenylmethylcarbamide C26HCH66-10)
[0204] The product from example 1 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml). 4-Fluorobenzaldehyde (0.08 ml, 0.6 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. IR: 1640, 1175, 1 1 10, 805, 700, 620 cm"1; LC- MS: (M+H)+ 446.3, tr 5.62 min.
Example 9 - N-f(4-Methylphenyl)methylVN-π-fphenylmethvDpiperidin-4-yr)-N'- phenylmeth ylcarbamide hydrochloride (26HCH16D)
[0205] To a solution of l-benzyl-4-piperidone (1.74 g, 9.2 mmol) and 4- methylbenzylamine (0.97 g, 8 mmol) in methanol (30 ml) was added sodium borohydride (525 mg) in small portions over 30 min. The reaction mixture was stirred at room temperature. After 16 h, the mixture was concentrated. Water (30 ml) was added, and the mixture was extracted with dichloromethane (2 X 20 ml). The combined organic layers were dried (Na2SO4), filtered, and concentrated to give 4-((4- methylphenyl)methyl)amino-l-phenylmethylpiperidine. The crude product was used without further purification.
[0206] 4-((4-Methylphenyl)methyl)amino-l-phenylmethylpiperidine (800 mg, 2.7 mmol) was dissolved in dry dichloromethane (30 ml). Benzylisocyanate (543 mg, 4.1 mmol) was added. The reaction mixture was stirred at room temperature. After 16 h, water (10 ml) was added followed by NaOH (6 N, 2 ml). After additional 30 minutes of stirring the white crystals were filtered off. The organic layer was isolated and dried (Na2SO4), filtered, and concentrated. Flash chromatography in dichloromethane/methanol 10/1 left N-((4-Methylphenyl)methyl)-N-( 1 -(phenylmethyl)piperidin-4-yl)-N'-phenylmet hylcarbamide Yield: 820 mg, 71%; A sample was concentrated with HCl (4 M in dioxane)followed by recrystallization from dichloromethane/diethyl ether leaving the title compound. 1H-NMR (CDCl3): δ 1.87 (brd, 2 H), 2.30 (s, 3 H)3 2.59 (dq, 2 H), 2.76 (br q, 2 H), 3.44 (br d, 2 H), 4.09 (d, 2 H), 4.30 (d, 2 H), 4.40 (s, 2 H), 4.64-4.76 (m, 2 H)5 6.98- 7.64 (Aromatic protons, 14 H); 13C-NMR (CDCl3): δ 21.2, 26.7, 45.0, 46.0, 49.7, 52.2, 61.0, 126.2, 127.26, 126.31, 128.2, 128.6, 129.6, 129.9, 130.4, 131.6, 134.4, 137.6, 139.3, 158.5; 13C-NMR (CD3OD5 rotamers): δ 19.8, 26.4, 27.8, 40.3, 44.3, 51.6, 51.9, 54.5, 60.5, 110.0, 112.1, 114.0, 114.2, 117.5, 125.9, 126.2, 126.7, 126.8, 128.9, 129.1, 129.2, 129.4, 129.7, 130.1, 131.2, 134.5, 137.4, 159.1, 173.8, 175.0; Mp. 109-112° C; Elemental analysis: Found C, 70.06; H, 7.62; N, 8.60; calcd for monohydrate: C, 69.76; H, 7.53; N, 8.72.
Example 10 - N-(f 4-MemylphenyDmethy IVN-O -fphenylmethv0piperidin-4-yl)-N'- phenylmeth ylcarbamide oxalate (34JJ59oxaD
[0207] N-((4-Methylphenyl)methyl)-N-( 1 -(phenylmethyl)piperidin-4-yl)-N'- phenylmeth ylcarbamide was prepared as described in example 9 above. A sample was precipitated as the oxalate and recrystallized from ethyl acetate to give the title compound. 13C-NMR (CDCl3): δ 21.2, 27.0, 45.0, 45.9, 49.9, 52.1, 60.6, 126.1, 127.3, 127.4, 128.5, 128.7, 129.6, 130.0, 130.4, 131.2, 134.3, 137.7, 139.3, 158.4, 163.4; Mp. 180-182° C; Elemental analysis: Found C, 69.54; H, 6.73; N, 7.96; calcd for monooxalate: C, 69.61; H, 6.82; N, 8.12.
Example 11 - N-r^-MethylphenvDmethylVN-π-Cphenylmethyπpiperidin^-yl)^- methoxyphen ylacetamide hydrochloride (26HCH17)
[0208] To a solution of l-benzyl-4-piperidone (1.74 g, 9.2 mmol) and 4- methylbenzylamine (0.97 g, 8 mmol) in methanol (30 ml) was added sodium borohydride (525 mg) in small portions over 30 min. The reaction mixture was stirred at room temperature. After 16 h, the mixture was concentrated. Water (30 ml) was added, and the mixture was extracted with dichloromethane (2 X 20 ml). The combined organic layers were dried (Na2SO4), filtered, and concentrated to give 4-((4- methylphenyl)methyl)amino-l-phenylmethylpiperidine. The crude product was used without further purification.
[0209] To a solution of 4-((4-Methylphenyl)methyl)arnino-l- phenylmethylpiperidine (800 mg, 2.7 mmol) in dry dichloromethane (30 ml) was added diisopropylethylamine (1.5 ml)followed by 4-methoxyphenylacetyl chloride (997 mg, 5.4 mmol). The reaction mixture was stirred at room temperature. After 16 h, the reaction mixture was concentrated, redissolved in diethyl ether, and extracted with HCl (0.6 N). The aqueous layer was isolated, treated with NaOH (1 N) until basic, and extracted with dichloromethane (20 ml). The organic layer was isolated and dried (Na2SO4), filtered, and concentrated, and redissolved in diethyl ether. The hydrochloride was formed by addition of HCl (4 M in dioxane), and recrystallized from diethyl ether to give the title compound. Yield: 600 mg, 50%; 1H-NMR (CDCl3): δ 1.75 (d, 2 H), 2.32 (s, 3 H), 2.50 (q, 2 H), 2.70 (q, 2 H), 3.38 (d, 2 H), 3.54 (s, 2 H)5 3.78 (s, 3 H), 4.06 (d, 2 H), 4.54 (s, 2 H), 4.82 (m, 1 H), 6.78-7.60 (aromatic protons, 13 H); 13C-NMR (CDCl3): δ 21.0, 26.0, 40.3, 46.3, 49.0, 51.8, 55.3, 60.8, 114.2, 125.6, 126.6, 127.9, 129.4, 129.60, 129.62, 130.3, 131.4, 134.8, 137.1, 158.7, 172.9; Mp. 197-200° C; Elemental analysis: Found C, 71.29; H, 7.25; N, 5.73; calcd for hydrate: C, 71.37; H, 7.43; N, 5.74.
Example 12 - N-((4-Methylphenyl)methyl)-N-fl-(phenylmethyl)piperidin-4-vπ-4- methoxyphen ylacetamide oxalate (34JJ61oxal)
10210] N-((4-Methylphenyl)methyl)-N-(l-(phenylmethyl)piperidin-4-yl)-4- methoxyphen ylacetamide was prepared as described in example 11 above. A sample was precipitated as the oxalate and recrystallized from tetrahydrofuran to give the title compound. 13C-NMR (CDCl3): δ 21.2, 26.4, 40.6, 52.0, 55.5, 114.4, 125.9, 126.7, 128.4, 129.6, 129.8, 129.9, 130.4, 131.2, 134.6, 137.6, 158.9, 163.3, 172.9; Mp. 171-173° C; Elemental analysis: Found C, 69.56; H, 6.74; N, 5.16; calcd for monooxalate: C, 69.48; H, 6.61; N, 5.40.
Example 13 - N-rr4-Methylphenyl)methyl)-N-(piperidin-4-yl)-4-methoxyphenylacetamide C26HCH71B)
[0211] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (1.75 g, 8.8 mmol) and 4-methylben2ylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (1 M, 6.7 ml) followed by NaCNBH3 in methanol (0.3 M, 30 ml). The resulting solution was stirred at room temperature. After 20 h, water (5 ml) was added, and the mixture was stirred for 1 h before it was concentrated. Flash chromatography in dichloromethane :methanol 10:1 gave tert-butyl 4- (4-methylphenyl)methyl)amino-piperidine carboxylate. Yield: 2.4 g, 98%. To a solution of tert-butyl 4-(4-methylphenyl)methyl)amino-piperidine carboxylate (862 mg, 2.83 mmol) in dry dichloromethane (10 ml) was added diisopropylethylamine (1.1 ml, 6.5 mmol) followed by 4-methoxyphenylacetyl chloride (0.66 ml, 4.3 mmol). The reaction mixture was stirred at room temperature. After 48 h, water (5 ml) was added, and the mixture was stirred for additional 2 h before extracted with NaOH (0.2 N, 2 X 15 ml), HCl (0.2 N5 2 X 15 ml), and water (15 ml). The organic layer was dried (Na2SO4) and concentrated to give N-((4-methylphenyl)methyl)-N~(l-(tert-butyloxycarbonyl)piperidin- 4-yl)-4-methoxyphenylacetamide. The crude product was used without any further purification. N-((4-Methylphenyl)methyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyphenylacetamide was dissolved in ether (2 ml) and HCl (3 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2 h, water (5 ml) was added, and the mixture was extracted with HCl (0.1 N, 3 X 30 ml). The combined aqueous layers were treated with NaOH (0.2 N) until basic (pH >8). The aqueous layer was extracted with diethyl ether (2 X 20 ml). The combined organic layers were dried (Na2SO4) and concentrated, before dissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated. Additional flash chromatography in dichloromethane/methanol 1/l.fwdarw.methanol containing 2% NH3 gave the title compound. Yield: 466 mg, 47%; 13C-NMR (CD3OD3 rotamers): δ 19.9, 27.8, 29.7, 40.2, 40.3, 44.4, 44.45, 44.50, 52.4, 54.5, 55.5, 114.0, 114.1, 126.0, 126.7, 126.9, 127.3, 128.7, 129.3, 129.6, 129.7, 135.1, 136.1, 136.2, 137.1, 159.0, 159.1, 173.1, 173.7. .
Example 14 - N-π-r3.3-Dimethylbutvnpiperidin-4-yl')-N-^4-methylphenvnmethvn-4- methox vphenylacetamide (26HCH79-5)
[0212] The product from example 13 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml). 3,3-Dimethylbutyraldehyde (0.143 ml, 1.1 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 26 mg; 13C-NMR (CD3OD, rotamers): δ 19.9, 27.4, 28.4, 28.8, 29.2. 29.3, 38.3, 38.4, 40.2, 40.3, 44.3, 52.0, 52.3, 52.4, 53.9, 54.6, 54.9, 114.0, 114.1, 126.0, 126.8, 127.0, 127.3, 128.8, 129.4, 129.8, 129.9, 135.0, 136.1, 136.3, 137.1, 158.96, 159.05, 173.2, 173.8.
Example 15 - N-ri-(Cvclohexylmethyl)piperidin-4-yπ-N-(C4-methylphenyl)methvπ-4- methoxy phenylacetamide (26HCH79-6)
[0213] The product from example 13 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml). Cyclohexanecarboxaldehyde (0.138 ml, 1.1 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 17 mg; HRMS (FAB+, NBA) (M+H)+ 449.3163, C29H4IN2O2 requires 449.3168; LC-MS: (M+H)+ 449.2, tr 7.92 min.
Example 16 - N-f(4-MemylphenyDmethyO-N-π-(2-methylpropyl)piperidin-4-yl)-4- methoxyph enylacetamide (26HCH79-7)
[0214] The product from example 13 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml).). 2-Methylpropionaldehyde (0.104 ml, 1.1 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 19 mg; HRMS (FAB+ , NBA) (M+H)+ 09.2858, C26H37N2O2 requires 409.2855; LC-MS: (M+H)+ 409.2, tr 5.97 min.
Example 17 - N-((4-Methylphenyl)methyl')-N-π-('f4-methylphenvπmethyl')piperidin-4- ylV4-m ethoxyphenylacetamide (26HCH79-8")
[0215] The product from example 13 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml). 4-Methylbenzaldehyde (0.134 ml, 1.1 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 22 mg; HRMS (FAB+, NBA) (M+H)+ 457.2853, C30H37N2O2 requires 457.2855; LC-MS: (MH-H)+ 457.2, tr 6.97 min.
Example 18 - N-f 1 -f (4-HvdroxyphenvDmethvDpiperidin-4-ylVN-(Y 4- methylphenyl)methylV4- methoxyphenylacetamide (26ΑCWJ9-9)
[0216] The product from example 13 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml). 4-Hydroxybenzaldehyde (139 mg, 1.1 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 19 mg; HRMS (FAB+, NBA) (M+H)+ 459.2655, C29H35N2O3 requires 459.2648; LC-MS: (M+H)+ 459.2, tr 2.84 min.
Example 19 z N-d-r(2-Hvdroxyphenvnmethvnpiperidin-4-yl)-N-((4- methylphenvDmethvD-4- methoxyphenylacetamide f26HCH79-10^
[0217] The product from example 13 above (20 mg, 0.06 mmol) was dissolved in abs. ethanol (2 ml). 2-Hydroxybenzaldehyde (0.122 ml, 1.1 mmol) was added followed by solid-supported borohydride (150 mg, 2.5 mmol/g resin; Aldrich 32,864-2). The mixture was shaken at room temperature. After 48 h, the resin was filtered off and acetic anhydride (0.02 ml, 0.2 mmol) was added to the organic solution. After 24 h, the mixture was concentrated and redissolved in methanol (2 ml). The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 16 mg; HRMS (FAB+, NBA) (MR-H)+ 459.2633, C29H35N2O3 requires 459.2648; LC-MS: (M+H)+ 459.2, tr 5.81 min.
Example 20 - N-(3-Phenylpropyl')-N-rpiperidin-4-yl)-4-methoxyphenylacetamide f26HCH80-n
[0218] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (400 mg, 2 mmol) in methanol (1 ml) and 3-phenylpropylamine (0.143 ml, 1 mmol) in methanol (1 ml) was added acetic acid in methanol (1 M, 1.34 ml) followed by NaCNBH3 in methanol (0.3 M, 4.4 ml). The resulting solution was stirred at room temperature. After 24 h, water (2 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. The resulting oil was redissolved in diethyl ether (20 ml), extracted with HCl (0.1 N3 1 X 15 ml). The aqueous layer was washed with diethyl ether (10 ml) and treated with 0.2 N NaOH until basic (pH>8), before extracted with dichloromethane (20 ml). The organic layer was dried (Na2SO4), filtered, and concentrated to give tert-butyl 4-(3-phenylpropyl)amino-piperidine carboxylate. Yield: 110 mg. To a solution of tert-butyl 4-(3-phenylpropyl)amino-piperidine carboxylate (50 mg, 0.16 mmol) in dichloromethane (6 ml) was added diisopropylethylamine (0.070 ml, 0.4 mmol) followed by 4-methoxyphenylacetyl chloride (0.055 ml, 0.35 mmol). The reaction mixture was stirred at room temperature. After 18 h, water (2 ml) was added. The mixture was stirred for another 2 h. The mixture was sequentially washed with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and water (10 ml), dried (Na2SO4), filtered and concentrated to give N-(3-phenylpropyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyph enylacetamide. The crude product was used without any further purification. N-(3-Phenylρropyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4-methoxyph enylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na2SO-O, filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 61 mg; 13C-NMR (CD3OD, rotamers): δ 27.8, 29.4, 30.8, 32.3, 32.7, 33.3, 40.2, 40.5, 42.0, 44.5, 44.6, 44.9, 52.7, 54.56, 54.57, 54.9, 114.0, 114.1, 125.7, 126.1, 127.0, 127.4, 128.2, 128.3, 128.5, 129.47, 129.55, 141.2, 141.8, 158.9, 159.0, 172.5, 172.7.
Example 21 - N-(2-Phenylethvπ-N-(pmeridin-4-vπ-4-metlioxyphenylmethylacetami(ie (26HCH80-2)
[0219] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (400 mg, 2 mmol) in methanol (1 ml) and 2-phenylethylamine (0.143 ml, 1 mmol) in methanol (1 ml) was added acetic acid in methanol (1 M, 1.34 ml) followed by NaCNBH3 in methanol (0.3 M, 4.4 ml). The resulting solution was stirred at room temperature. After 24 h, water (2 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. The resulting oil was redissolved in diethyl ether (20 ml), extracted with HCl (0.1 N, 1 X 15 ml). The aqueous layer was washed with diethyl ether (10 ml) and treated with 0.2 N NaOH until basic (ρH>8), before extracted with dichloromethane (20 ml). The organic layer was dried (Na2SO4), filtered, and concentrated to give tert-butyl 4-(2-phenylethyl)amino-piperidine carboxylate. Yield: 221 mg. To a solution of tert-butyl 4-(2-phenylethyl)amino-piperidine carboxylate (50 mg, 0.16 mmol) in dichloromethane (6 ml) was added diisopropylethylamine (0.070 ml, 0.4 mmol) followed by 4-methoxyphenylacetyl chloride (0.055 ml, 0.35 mmol). The reaction mixture was stirred at room temperature. After 18 h, water (2 ml) was added. The mixture was stirred for another 2 h. The mixture was sequentially washed with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and water (10 ml), dried (Na2SO4), filtered and concentrated to give N-(2-phenylethyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyphe nylacetamide. The crude product was used without any further purification. N-(2-Phenylethyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4-methoxyphe nylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na2SO4), filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 52 mg; 13C-NMR (CD3OD5 rotamers): δ 27.1, 28.5, 34.9, 36.6, 40.2, 40.4, 44.1, 44.2, 44.4, 53.3, 54.2, 54.6, 114.0, 114.1, 126.2, 126.6, 127.2, 127.4, 128.3, 128.6, 128.79, 128.82, 129.7, 138.5, 139.5, 158.96, 159.0,
172.7, 173.1
Example 22 : N-f(2-Methoxyphenyl')methyiyN-fpiperidin-4-yl)-4- methoxyphenylmethylacetami de C26HCH80-4)
[0220] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (400 mg, 2 mmol) in methanol (1 ml) and 2-methoxybenzylamine (0.130 ml, 1 mmol) in methanol (1 ml) was added acetic acid in methanol (1 M, 1.34 ml) followed by NaCNBHb in methanol (0.3 M, 4.4 ml). The resulting solution was stirred at room temperature. After 24 h, water (2 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. The resulting oil was redissolved in diethyl ether (20 ml), extracted with HCl (0.1 N, 1 X 15 ml). The aqueous layer was washed with diethyl ether (10 ml) and treated with 0.2 N NaOH until basic (pH>8), before extracted with dichloromethane (20 ml). The organic layer was dried (Na2SO4), filtered, and concentrated to give tert-butyl 4-((2-methoxyphenyl)methyl)amino-piperidine carboxylate. Yield: 211 mg. To a solution of tert-butyl 4-((2- methoxyphenyl)methyl)amino-piperidine carboxylate (50 mg, 0.16 mmol) in dichloromethane (6 ml) was added diisopropylethylamine (0.070 ml, 0.4 mmol) followed by 4-methoxyphenylacetyl chloride (0.055 ml, 0.35 mmol). The reaction mixture was stirred at room temperature. After 18 h, water (2 ml) was added. The mixture was stirred for another 2 h. The mixture was sequentially washed with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and water (10 ml), dried (Na2SO4), filtered and concentrated to give N-((2-methoxyphenyl)methyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyphenylacetamide. The crude product was used without any further purification. N-((2-Methoxyphenyl)methyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na2SO4), filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 40 mg; 13C-NMR (CD3OD, rotamers): δ 26.1, 27.4, 40.0, 40.1, 43.5, 43.9, 51.5, 53.4, 54.5, 54.58, 54.63, 54.78, 54.83, 1 10.1, 110.5, 113.76, 113.78, 113.84, 114.0, 114.1, 120.1, 120.5, 125.4, 126.0, 126.5, 126.7, 127.1, 127.3, 127.7, 128.8, 129.8, 130.0, 130.08, 130.14, 156.5, 157.0, 159.0, 159.1, 173.2, 173.8.
Example 23 - N-(('2-Chlorophenyl')methyl')-N-('piperidin-4-vπ-4-niethoxyphenylacetainide (26HCH80-5^
[0221] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (400 mg, 2 mmol) in methanol (1 ml) and 2-chlorobenzylamine (0.121 ml, 1 mmol) in methanol (1 ml) was added acetic acid in methanol (1 M, 1.34 ml) followed by NaCNBhL) in methanol (0.3 M, 4.4 ml). The resulting solution was stirred at room temperature. After 24 h, water (2 ml) was added, and the mixture was stirred for 1 h before it was concentrated. The resulting oil was redissolved in diethyl ether (20 ml), extracted with HCl (0.1 N, 1 X 15 ml). The aqueous layer was washed with diethyl ether (10 ml) and treated with 0.2 N NaOH until basic (pH>8), before extracted with dichloromethane (20 ml). The organic layer was dried (Na2SO-t), filtered, and concentrated to give tert-butyl 4-((2-chlorophenyl)methyl)amino-piperidine carboxylate. Yield: 137 mg. To a solution of tert-butyl 4-((2-chlorophenyl)rnethyl)amino-piperidine carboxylate (50 mg, 0.15 mmol) in dichloromethane (6 ml) was added diisopropylethylamine (0.070 ml, 0.4 mmol) followed by 4-methoxyphenylacetyl chloride (0.055 ml, 0.35 mmol). The reaction mixture was stirred at room temperature. After 18 h, water (2 ml) was added. The mixture was stirred for another 2 h. The mixture was sequentially washed with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and water (10 ml), dried (Na2SO4), filtered and concentrated to give N-((2-chlorophenyl)methy I)-N- (l-(tert-butyloxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide. The crude product was used without any further purification. N-((2-Chlorophenyl)methyl)-N-(l-(tert- butyloxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na2SO4), filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 45 mg; 13C-NMR (CD3OD, rotamers): δ 25.8, 26.9, 40.0, 40.1, 42.9, 43.4, 43.7, 46.0, 51.1, 53.0, 54.6, 113.77, 113.84, 114.0, 114.1, 126.6, 126.8, 127.08, 127.13, 127.3, 127.4, 128.1, 129.0, 129.2, 129.8, 130.0, 130.2, 131.9, 132.2, 135.0, 135.3, 159.1, 173.4, 173.8.
Example 24. r N-((3.4-Di-methoxyphenyl')methyl)-N-rpiperidin-4-yl')-4- methoxyphenylacetamid e (26HCH80-6)
[0222] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (400 mg, 2 mmol) in methanol (1 ml) and 3,4-di-methoxybenzylamine (0.151 ml, 1 mmol) in methanol (1 ml) was added acetic acid in methanol (1 M, 1.34 ml) followed by NaCNBEb in methanol (0.3 M, 4.4 ml). The resulting solution was stirred at room temperature. After 24 h, water (2 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. The resulting oil was redissolved in diethyl ether (20 ml), extracted with HCl (0.1 N3 1 X 15 ml). The aqueous layer was washed with diethyl ether (10 ml) and treated with 0.2 N NaOH until basic (pH>8), before extracted with dichloromethane (20 ml). The organic layer was dried (Na2SO4), filtered, and concentrated to give tert-butyl 4-((3,4-di-methoxyphenyl)methyl)amino-piperidine carboxylate. Yield: 162 mg. To a solution of tert-butyl 4-((3,4-di- methoxyphenyl)methyl)amino-piperidine carboxylate (50 mg, 0.14 mmol) in dichloromethane (6 ml) was added diisopropylethylamine (0.070 ml, 0.4 mmol) followed by 4-methoxyphenylacetyl chloride (0.055 ml, 0.35 mmol). The reaction mixture was stirred at room temperature. After 18 h, water (2 ml) was added. The mixture was stirred for another 2 h. The mixture was sequentially washed with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and water (10 ml), dried (Na2SO4), filtered and concentrated to give N-((3 ,4-di-methoxyphenyl)methyl)-N-( 1 -(tert-butyloxycarbonyi)piperidin-4-yl)-4- methoxyphenylacetamide. The crude product was used without any further purification. N-((3 ,4-Di-methoxyphenyl)methyl)-N-( 1 -(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na2SO4), filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 54 mg; 13C-NMR (CD3OD, rotamers): δ 25.9, 27.3, 40.0, 40.1, 43.5, 43.8, 44.1, 51.4, 53.5, 54.6, 55.4, 110.2, 111.0, 111.9, 112.2, 114.0, 114.2, 118.6, 119.4, 127.1, 127.4, 129.9, 130.0, 130.5, 132.1, 148.2, 148.7, 149.2, 149.7, 158.98, 159.05, 173.3, 173.6.
Example 25 - N-((4-Fluorophenvπmethyl')-N-('piperidin-4-ylV4-methoxyphenylacetamide (26HCHSO^
[0223] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (400 mg, 2 mmol) in methanol (1 ml) and 4-fluorobenzylamine (0.114 ml, 1 mmol) in methanol (1 ml) was added acetic acid in methanol (1 M, 1.34 ml) followed by NaCNBH3 in methanol (0.3 M, 4.4 ml). The resulting solution was stirred at room temperature. After 24 h, water (2 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. The resulting oil was redissolved in diethyl ether (20 ml), extracted with HCl (0.1 N, 1 X 15 ml). The aqueous layer was washed with diethyl ether (10 ml) and treated with 0.2 N NaOH until basic (pH>8), before extracted with dichioromethane (20 ml). The organic layer was dried (Na2SO4), filtered, and concentrated to give tert-butyl 4-((4-fluorophenyl)rnethyl)amino-piperidine carboxylate. Yield: 130 mg. To a solution of tert-butyl 4-((4-fluorophenyl)methyl)amino-piperidine carboxylate (50 mg, 0.16 mmol) in dichioromethane (6 ml) was added diisopropylethylamine (0.070 ml, 0.4 mmol) followed by 4-methoxyphenylacetyl chloride (0.055 ml, 0.35 mmol). The reaction mixture was stirred at room temperature. After 18 h, water (2 ml) was added. The mixture was stirred for another 2 h. The mixture was sequentially washed with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and water (10 ml), dried (Na2SO4), filtered and concentrated to give N-((4-fluorophenyl)methy I)-N- (l-(tert-butyloxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide. The crude product was used without any further purification. N-((4-Fluorophenyl)methyl)-N-(l-(tert- butyloxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichioromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na2SO4), filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 45 mg; 13C-NMR (CD3OD, rotamers): δ 26.1, 27.5, 40.1, 43.6, 43.8, 44.0, 51.6, 53.6, 54.6, 113.77, 113.84, 114.0, 114.1, 114.7, 114.9, 115.3, 115.6, 126.8, 127.2, 128.1, 128.6, 128.7, 129.8, 130.0, 130.1, 130.6, 131.0, 133.8, 159.1, 173.3, 173.6.
Example 26. = N-ff2.4-Di-chlorophenvDmethyl)-N-Cpiperidin-4-vD-4- methoxyphenylacetamide f26HCH80-8)
[0224] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (400 mg, 2 mmol) in methanol (1 ml) and 2,4-di-chlorobenzylamine (0.135 ml, 1 mmol) in methanol (1 ml) was added acetic acid in methanol (1 M, 1.34 ml) followed by NaCNBH3 in methanol (0.3 M, 4.4 ml). The resulting solution was stirred at room temperature. After 24 h, water (2 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. The resulting oil was redissolved in diethyl ether (20 ml), extracted with HCl (0.1 N, 1 X 15 ml). The aqueous layer was washed with diethyl ether (10 ml) and treated with 0.2 N NaOH until basic (pH>8), before extracted with dichloromethane (20 ml). The organic layer was dried (Na2SO4), filtered, and concentrated to give tert-butyl 4-((2,4-di-chlorophenyl)methyl)amino-piperidine carboxylate. Yield: 97 mg. To a solution of tert-butyl 4-((2,4-di- chlorophenyl)methyl)amino-piperidine carboxylate (50 mg, 0.14 mmol) in dichloromethane (6 ml) was added diisopropylethylamine (0.070 ml, 0.4 mmol) followed by 4-methoxyphenylacetyl chloride (0.055 ml, 0.35 mmol). The reaction mixture was stirred at room temperature. After 18 h, water (2 ml) was added. The mixture was stirred for another 2 h. The mixture was sequentially washed with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and water (10 ml), dried (Na2SO4), filtered and concentrated to give N-((2,4-di-chlorophenyl)methyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyphenylacetamide. The crude product was used without any further purification. N-((2,4-Di-chlorophenyl)methyl)-N-(l-(tert-butyloxycarbonyl)piperidin-4-yl)-4- methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml. 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15. ml), dried (Na2SO4), filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 39 mg; 13C-NMR (CD3OD5 rotamers): δ 25.7, 26.8, 40.0, 42.6, 43.3, 43.7, 51.2, 53.0, 54.5, 54.6, 113.8, 113.8, 114.0, 1 14.1, 127.0, 128.4, 128.8, 129.8, 130.0, 130.1, 131.0, 132.7, 132.9, 134.5, 159.1, 173.4, 173.6.
Example 27 - N-(T3-MethylphenvQmethylVN-(piperidin-4-ylV4-methoxyphenvIacetamide (26HCHSO^
[0225] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (400 mg, 2 mmol) in methanol (1 ml) and 3-methylbenzylamine (0.125 ml, 1 mmol) in methanol (1 ml) was added acetic acid in methanol (1 M, 1.34 ml) followed by NaCNBH3 in methanol (0.3 M, 4.4 ml). The resulting solution was stirred at room temperature. After 24 h, water (2 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. The resulting oil was redissolved in diethyl ether (20 ml), extracted with HCl (0.1 N, 1 X 15 ml). The aqueous layer was washed with diethyl ether (10 ml) and treated with 0.2 N NaOH until basic (pH>8), before extracted with dichloromethane (20 ml). The organic layer was dried (Na2SO4), filtered, and concentrated to give tert-butyl 4-((3-methylphenyl)methyl)amino-piperidine carboxylate. Yield: 136 mg. To a solution of tert-butyl 4-((3-methylphenyl)methyl)amino-piperidine carboxylate (50 mg, 0.16 mmol) in dichloromethane (6 ml) was added diisopropylethylamine (0.070 ml, 0.4 mmol) followed by 4-methoxyphenylacetyl chloride (0.055 ml, 0.35 mmol). The reaction mixture was stirred at room temperature. After 18 h, water (2 ml) was added. The mixture was stirred for another 2 h. The mixture was sequentially washed with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and water (10 ml), dried (Na2SO4), filtered and concentrated to give N-((3 -methylpheny l)methy I)-N- (1 -(tert-butyl oxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide. The crude product was used without any further purification. N-((3-Methylphenyl)methyl)-N-(l-(tert- butyloxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na2SO4), filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 48 mg; 13C-NMR (CD3OD, rotamers): δ 20.4, 26.8, 28.3, 40.2, 43.9, 44.1, 44.5, 51.8, 54.2, 54.57, 54.61, 114.0, 114.1, 123.2, 123.7, 126.7, 127.0, 127.1, 127.3, 128.0, 128.1, 128.7, 129.8, 129.9, 137.9, 138.6, 138.9, 159.0, 159.1, 173.1, 173.7.
Example 28 - N-((3-Bromophenvnmethyl)-N-('piperidin-4-yl")-4-methoxyphenylacetaniide (26HCH80-10)
[0226] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (400 mg, 2 mmol) in methanol (1 ml) and 3-bromobenzylamine hydrobromide (222 mg, 1 mmol) in methanol (1 ml) was added acetic acid in methanol (1 M, 1.34 ml) followed by NaCNBH3 in methanol (0.3 M, 4.4 ml). The resulting solution was stirred at room temperature. After 24 h, water (2 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. The resulting oil was redissolved in diethyl ether (20 ml), extracted with HCl (0.1 N5 I X 15 ml). The aqueous layer was washed with diethyl ether (10 ml) and treated with 0.2 N NaOH until basic (pH>8), before extracted with dichloromethane (20 ml). The organic layer was dried (NaaSC^), filtered, and concentrated to give tert-butyl 4-((3-bromophenyl)methyl)amino-piperidine carboxylate. Yield: 142 mg. To a solution of tert-butyl 4-((3-bromophenyl)methyl)amino-piperidine carboxylate (50 mg, 0.14 mmol) in dichloromethane (6 ml) was added diisopropylethylamine (0.070 ml, 0.4 mmol) followed by 4-methoxyphenylacetyl chloride (0.055 ml, 0.35 mmol). The reaction mixture was stirred at room temperature. After 18 h, water (2 ml) was added. The mixture was stirred for another 2 h. The mixture was sequentially washed with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and water (10 ml), dried (Na2SO4), filtered and concentrated to give N-((3-bromophenyl)methyl)-N- (l-(tert-butyloxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide. The crude product was used without any further purification. N-((3-Bromophenyl)methyl)-N-(l-(tert- butyloxycarbonyl)piperidin-4-yl)-4-methoxyphenylacetamide was dissolved in diethyl ether (2 ml) and HCl (1 ml, 4 M in dioxane) was added. The reaction mixture was stirred at room temperature. After 2.5 h, NaOH (1 ml, 6 N) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 15 ml), dried (Na2SO4), filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 49 mg; 13C-NMR (CD3OD, rotamers): δ 26.6, 28.2, 40.2, 43.9, 44.0, 51.8, 54.1, 54.6, 113.76, 113.84, 114.1, 114.2, 122.2, 125.0, 125.5, 126.7, 127.1, 129.2, 129.5, 129.7, 129.8, 129.9, 130.0, 130.5, 130.6, 140.8, 141.8, 159.1, 173.3, 173.7.
Example 29 - N-(l-(Phenylmethvπpiperidin-4-ylVNr('3-phenyl-2-propen-l-vπ-4- methoxyphen ylacetamide C26HCH76B')
[0227] To a solution of 4-amino-N-benzylpiperidine (200 mg, 1.05 mmol) in methanol (2 ml) was added trans-cinnamaldehyde (211 mg, 1.6 mmol), followed by Acetic acid in methanol (1 M, 1.4 ml) and sodiumcyanoborohydride in methanol (0.3 M, 4.4 ml). The reaction mixture was stirred at room temperature. After 48 h, water (2 ml) was added. The mixture was stirred for another 2 h before concentrated and redissolved in diethyl ether (20 ml). The organic layer was extracted with HCl (0.1 N, 2 X 10 ml). The combined aqueous layers were treated with NaOH (0.2 N) until basic (pH>8). The mixture was extracted with dichloromethane (2 X 10 ml). The combined organic layers were dried (Na2SO4) and concentrated. The crude product, which was used without any further purification, was dissolved in dichloromethane (5 ml). Diisopropylethylamine (284 mg, 2.1 eq.) was added, followed by 4-methoxyphenylacetyl chloride (387 mg, 2.0 eq). The reaction mixture was stirred at room temperature. After 18 h, water (2 ml) was added. After additional 2 h dichloromethane (10 ml) was added. The mixture was extracted with NaOH (0.2 N, 3 X 15 ml), and water (15 ml). The organic layer was dried (Na2SO4) and concentrated. The crude product was redissolved in methanol (2 ml) and added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. '3C-NMR (CDCl3): δ 28.5, 38.1, 46.6, 47.4, 50.9, 54.7, 62.9, 113.7, 125.5, 126.4, 126.6, 127.4, 127.9, 128.5, 128.6, 129.6, 130.0, 135.2, 135.3, 138.O5 158.2, 173.2.
Example 30 - N-f(4-MethyIphenyl)methyl)-N-n^iperidin-4-ylVphenylacetamide (26HCH78-n
[0228] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (1.75 g, 8.8 mmol) and 4-methylbenzylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (1 M, 6.7 ml) followed by NaCNBH3 in methanol (0.3 M, 30 ml). The resulting solution was stirred at room temperature. After 20 h, water (5 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. Flash chromatography in dichloromethane:methanol 10:1 gave tert-butyl 4- (4-methylphenyl)methyl)amino-piperidine carboxylate. Yield: 2.4 g, 98%. To a solution of tert-butyl 4-(4-methylphenyl)methyl)amino-piperidine carboxylate (80 mg, 0.26 mmol) in dichloromethane (1.8 ml) was added diisopropylethylamine (0.11 ml, 2.4 eq.) followed by phenylacetyl chloride (81 mg, 0.53 mmol). The reaction mixture was stirred at room temperature. After 20 h, water (1 ml) was added. The mixture was stirred for another 2 h, before diethyl ether (20 ml) was added. The mixture was sequentially extracted with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N5 2 X 15 ml), and H2O (10 ml), dried (Na2SO4), filtered and concentrated. The crude material was dissolved in diethyl ether (2 ml) and HCl (4 M in dioxane, 1 ml). The reaction mixture was stirred at room temperature. After 2 h, NaOH (6 N, 1 ml) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 10 ml), dried (Na2SO4), and filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 38 mg; 13C-NMR (CD3OD, rotamers): δ 19.9, 26.9, 28.4, 41.0, 41.1, 44.0, 44.1, 44.4, 51.9, 54.4, 126.1, 126.7, 126.8, 126.9, 128.5, 128.7, 128.78, 128.81, 128.9, 129.4, 129.5, 134.9, 135.2, 135.6, 136.0, 136.3, 137.2, 172.8, 173.3.
Example 31 - • N-rf4-Methylphenvπmethyl)-N-ri-piperidin-4-yl')-3-phenylpropionamide f26HCH78-2~)
[0229J To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (1.75 g,. 8.8 mmol) and 4-methylbenzylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (I M, 6.7 ml) followed by NaCNBH3 in methanol (0.3 M, 30 ml). The resulting solution was stirred at room temperature. After 20 h, water (5 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. Flash chromatography in dichloromethane :methanol 10:1 gave tert-butyl 4- (4-methylphenyl)methyl)amino-piperidine carboxylate. Yield: 2.4 g, 98%. To a solution of tert-butyl 4-(4-methylphenyl)methyl)amino-piperidine carboxylate (80 mg, 0.26 mmol) in dichloromethane (1.8 ml) was added diisopropylethylamine (0.11 ml, 2.4 eq.) followed by 3-phenylpropionyl chloride (0.078 ml, 0.53 mmol). The reaction mixture was stirred at room temperature. After 20 h, water (1 ml) was added. The mixture was stirred for another 2 h, before diethyl ether (20 ml) was added. The mixture was sequentially extracted with HCl (0.2 N. 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and H2O (10 ml), dried (Na2SO4), filtered and concentrated. The crude material was dissolved in diethyl ether (2 ml) and HCl (4 M in dioxane, 1 ml). The reaction mixture was stirred at room temperature. After 2 h, NaOH (6 N, 1 ml) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 10 ml), dried (Na2SO4), and filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 43 mg; 13C-NMR (CD3OD, rotamers): δ 19.9, 27.4, 29.0, 31.4, 31.7, 34.7, 35.7, 44.2, 44.3, 51.6, 54.2, 125.9, 126.07, 126.15, 126.8, 128.3, 128.4, 128.7, 128.8, 129.3, 135.1, 136.1, 136.2, 137.0, 141.1, 141.2, 173.9, 174.4.
Example 32 - N-((4-Methylphenyl')methyl*)-N-(l-piperidin-4-ylVfphenylthio')acetamide (26HCH78-31
[0230] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (1.75 g, 8.8 mmol) and 4-methylbenzylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (1 M, 6.7 ml) followed by NaCNBH3 in methanol (0.3 M, 30 ml). The resulting solution was stirred at room temperature. After 2O h, water (5 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. Flash chromatography in dichloromethane :methanol 10:1 gave tert-butyl 4- (4-methylphenyl)methyl)amino-piperidine carboxylate. Yield: 2.4 g, 98%. To a solution of tert-butyl 4-(4-methylphenyl)methyl)amino-piperidine carboxylate (80 mg, 0.26 mmol) in dichloromethane (1.8 ml) was added diisopropylethylamine (0.11 ml, 2.4 eq.) followed by (phenylthio)acetyl chloride (0.078 ml, 0.53 mmol). The reaction mixture was stirred at room temperature. After 20 h, water (1 ml) was added. The mixture was stirred for another 2 h, before diethyl ether (20 ml) was added. The mixture was sequentially extracted with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and H2O (10 ml), dried (Na2SO4), filtered and concentrated. The crude material was dissolved in diethyl ether (2 ml) and HCl (4 M in dioxane, 1 ml). The reaction mixture was stirred at room temperature. After 2 h, NaOH (6 N, 1 ml) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 10 ml), dried (Na2SO4), and filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 18 mg; HRMS (FAB+, NBA) (M+H)+ 355.1841, C2iH27N2OS requires 355.1844; LC-MS: (M+H)+ 355.1, tr 2.62 min.
Example 33 - N-rf4-MethylphenylΛ)methyl)-N-π-piperidin-4-yl)-phenoxyacetamide f26HCH78-4>
[0231] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (1.75 g, 8.8 mmol) and 4-methylbenzylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (1 M, 6.7 ml) followed by NaCNBH3 in methanol (0.3 M, 30 ml). The resulting solution was stirred at room temperature. After 20 h, water (5 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. Flash chromatography in dichloromethanermethanol 10:1 gave tert-butyl 4- (4-methylphenyl)methyl)amino-piperidine carboxylate. Yield: 2.4 g, 98%. To a solution of tert-butyl 4-(4-methylphenyl)methyl)amino-piperidine carboxylate (80 mg, 0.26 mmol) in dichloromethane (1.8 ml) was added diisopropylethylamine (0.11 ml, 2.4 eq.) followed by phenoxyacetyl chloride (0.073 ml, 0.53 mmol). The reaction mixture was stirred at room temperature. After 20 h, water (1 ml) was added. The mixture was stirred for another 2 h, before diethyl ether (20 ml) was added. The mixture was sequentially extracted with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N5 2 X 15 ml), and H2O (10 ml), dried (Na2SO4), filtered and concentrated. The crude material was dissolved in diethyl ether (2 ml) and HCl (4 M in dioxane, 1 ml). The reaction mixture was stirred at room temperature. After 2 h, NaOH (6 N, 1 ml) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 10 ml), dried (Na2SO4), and filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 24 mg; 13C-NMR (CD3OD, rotamers): δ 19.9, 25.8. 27.4, 43.5, 43.7, 44.4, 51.9, 52.3, 66.9, 114.7, 114.8, 116.7, 117.0, 121.4, 123.6, 126.3, 126.8, 128.4, 128.9, 129.3, 129.5, 129.6, 131.0, 134.4, 136.1, 137.4, 158.3, 169.8, 170.1.
Example 34 - N-((4-Methylphenyl>methyl)-N-π -piperidin-4-vn-f 4- chlorophenoxy")acetamide f26HCH78-5') [0232] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (1.75 g, 8.8 mmol) and 4-methylbenzylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (1 M, 6.7 ml) followed by NaCNBHa in methanol (0.3 M, 30 ml). The resulting solution was stirred at room temperature. After 20 h, water (5 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. Flash chromatography in dichloromethane:methanol 10:1 gave tert-butyl 4- (4-methylphenyl)methyl)amino-piperidine carboxylate. Yield: 2.4 g, 98%. To a solution of tert-butyl 4-(4-methylphenyl)rnethyl)amino-piperidine carboxylate (80 mg, 0.26 mmol) in dichloromethane (1.8 ml) was added diisopropylethylamine (0.11 ml, 2.4 eq.) followed by 4-chlorophenoxyacetyl chloride (0.082 ml, 0.53 mmol). The reaction mixture was stirred at room temperature. After 20 h, water (1 ml) was added. The mixture was stirred for another 2 h, before diethyl ether (20 ml) was added. The mixture was sequentially extracted with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and H2O (10 ml), dried (Na2SO4), filtered and concentrated. The crude material was dissolved in diethyl ether (2 ml) and HCl (4 M in dioxane, 1 ml). The reaction mixture was stirred at room temperature. After 2 h, NaOH (6 N, 1 ml) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 10 ml), dried (Na2SO4), and filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 21 mg; 13C-NMR (CD3OD3 rotamers): δ 19.9, 26.2, 27.8, 43.6, 43.9, 44.4, 52.2, 52.5, 67.0, 116.2, 116.4, 126.2, 126.3, 126.8, 128.6, 128.9, 129.1, 129.3, 129.5, 131.0, 134.4, 135.6, 136.4, 137.5, 157.1, 169.4, 169.7.
Example 3Jj - . N-C(4-Methylphenyl>methylVN-π-piperidin-4-vn-3- methoxyphenylacetamide (26HCH78-6)
[0233] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (1.75 g, 8.8 mmol) and 4-methylbenzylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (1 M, 6.7 ml) followed by NaCNBH3 in methanol (0.3 M5 30 ml). The resulting solution was stirred at room temperature. After 20 h, water (5 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. Flash chromatography in dichloromethane:methanol 10:1 gave tert-butyl 4- (4-methylphenyl)methyl)amino-piperidine carboxylate. Yield: 2.4 g, 98%. To a solution of tert-butyl 4-(4-methylphenyl)methyl)amino-piperidine carboxylate (80 mg, 0.26 mmol) in dichloromethane (1.8 ml) was added diisopropylethylamine (0.11 ml, 2.4 eq.) followed by 3-methoxyphenylacetyl chloride (97 mg, 0.53 mmol). The reaction mixture was stirred at room temperature. After 20 h, water (1 ml) was added. The mixture was stirred for another 2 h, before diethyl ether (20 ml) was added. The mixture was sequentially extracted with HCl (0.2 N5 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and H2O (10 ml), dried (Na2SO4), filtered and concentrated. The crude material was dissolved in diethyl ether (2 ml) and HCl (4 M in dioxane, 1 ml). The reaction mixture was stirred at room temperature. After 2 h, NaOH (6 N, 1 ml) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 10 ml), dried (Na2SO4), and filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 26 mg; 13C-NMR (CD3OD, rotamers): δ 19.9, 26.3, 27.7, 41.0, 43.7, 43.9, 44.4, 51.5, 53.8, 54.5, 54.6, 112.2, 112.6, 114.3, 114.5, 121.0, 121.2, 126.1, 126.8, 128.8, 129.4, 129.5, 129.8, 134.8, 136.0, 136.3, 136.5, 136.9, 137.2, 160.2, 160.3, 172.8, 173.2.
Example 36 - N-((4-Methylphenyl)methyl)-N-(l-piperidin-4-yl)-4-fluorophenylacetamide (26HCH78-7)
[0234] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (1.75 g, 8.8 mmol) and 4-methylbenzylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (1 M, 6.7 ml) followed by NaCNBH3 in methanol (0.3 M, 30 ml). The resulting solution was stirred at room temperature. After 20 h, water (5 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. Flash chromatography in dichloromethane rmethanol 10:1 gave tert-butyl 4- (4-methylphenyl)methyl)amino-piperidine carboxylate. Yield: 2.4 g, 98%. To a solution of tert-butyl 4-(4-methylphenyl)methyl)amino-piperidine carboxylate (80 mg, 0.26 mmol) in dichloromethane (1.8 ml) was added diisopropylethylamine (0.11 ml, 2.4 eq.) followed by 4-fiuorophenylacetyl chloride (0.072 ml, 0.53 mmbl). The reaction mixture was stirred at room temperature. After 20 h, water (1 ml) was added. The mixture was stirred for another 2 h, before diethyl ether (20 ml) was added. The mixture was sequentially extracted with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and H2O (10 ml), dried (Na2SO4), filtered and concentrated. The crude material was dissolved in diethyl ether (2 ml) and HCl (4 M in dioxane, 1 ml). The reaction mixture was stirred at room temperature. After 2 h, NaOH (6 N, 1 ml) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 10 ml), dried (Na2SO4), and filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 rnmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 26 mg; 13C-NMR (CD3OD5 rotamers): δ 19.9, 26.1, 27.4, 39.7, 39.9, 43.5, 43.8, 44.4, 51.3, 53.4, 114.9, 115.1, 115.3, 126.1, 126.7, 128.5, 128.8, 129.4, 130.7, 130.8, 130.9, 131.0, 131.2, 131.6, 134.8, 136.0, 136.3, 137.2, 160.9, 163.3, 172.7, 173.2.
Example 37 N-f f4-MethylphenyDmethvn-N-( 1 -piperidin-4-vD-2.5-di- methoxyphenylacetami de (26HCH78-8)
[0235] To a solution of commercially available tert-butyl 4-oxo-l-piperidine carboxylate (1.75 g, 8.8 mmol) and 4-methylbenzylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (1 M, 6.7 ml) followed by NaCNBH3 in methanol (0.3 M, 30 ml). The resulting solution was stirred at room temperature. After 20 h, water (5 ml) was added, and the mixture was stirred for 1 h5 before it was concentrated. Flash chromatography in dichloromethane :methanol 10:1 gave tert-butyl 4- (4-methylρhenyl)methyl)amino-piperidine carboxylate. Yield: 2.4 g, 98%. To a solution of tert-butyl 4-(4-methylphenyl)methyl)amino-piperidine carboxylate (80 mg, 0.26 mmol) in dichloromethane (1.8 ml) was added diisopropylethylamine (0.11 ml, 2.4 eq.) followed by 2,5-di-methoxyphenylacetyl chloride (0.092 ml, 0.53 mmol). The reaction mixture was stirred at room temperature. After 20 h, water (1 ml) was added. The mixture was stirred for another 2 h, before diethyl ether (20 ml) was added. The mixture was sequentially extracted with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and H2O (10 ml), dried (Na2SO4), filtered and concentrated. The crude material was dissolved in diethyl ether (2 ml) and HCl (4 M in dioxane, 1 ml). The reaction mixture was stirred at room temperature. After 2 h, NaOH (6 N, 1 ml) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 10 ml), dried (Na2SO4), and filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. 36 mg; 13C-NMR (CD3OD, rotamers): δ 20.0, 26.5, 28.2, 35.1, 35.7, 44.0, 44.4, 51.6, 53.8, 54.99, 55.03, 55.2, 55.5, 111.4, 111.7, 112.4, 112.9, 116.6, 116.9, 124.98, 125.02, 126.1, 126.7, 128.8, 129.3, 135.0, 136.1, 136.3, 137.0, 151.3, 151.7, 153.9, 154.0, 173.1, 173.5.
Example 38 - N-(Y4-MethylphenyDmethviyN-f l-piperidin-4-yl')-4-chlorophenylacetamide (26HCH78-9>>
[0236] To a solution of commercially available tert-butyl 4-oxo-l-piperidϊne carboxylate (1.75 g, 8.8 mmol) and 4-methylbenzylamine (970 mg, 8.0 mmol) in methanol (7 ml) was added acetic acid in methanol (I M, 6.7 ml) followed by NaCNBH3 in methanol (0.3 M, 30 ml). The resulting solution was stirred at room temperature. After 20 h, water (5 ml) was added, and the mixture was stirred for 1 h, before it was concentrated. Flash chromatography in dichloromethane:methanol 10: 1 gave tert-butyl 4- (4-methylphenyl)methyl)amino-piperidine carboxylate. Yield: 2.4 g, 98%. To a solution of tert-butyl 4-(4-methylphenyl)methyl)amino-piperidine carboxylate (80 mg, 0.26 mmol) in dichloromethane (1.8 ml) was added diisopropylethylamine (0.1 1 ml, 2.4 eq.) followed by 4-chlorophenylacetyl chloride (99 mg, 0.53 mmol). The reaction mixture was stirred at room temperature. After 20 h, water (1 ml) was added. The mixture was stirred for another 2 h, before diethyl ether (20 ml) was added. The mixture was sequentially extracted with HCl (0.2 N, 2 X 15 ml), NaOH (0.2 N, 2 X 15 ml), and H2O (10 ml), dried (Na2SO4), filtered and concentrated. The crude material was dissolved in diethyl ether (2 ml) and HCl (4 M in dioxane, 1 ml). The reaction mixture was stirred at room temperature. After 2 h, NaOH (6 N, 1 ml) was added followed by dichloromethane (10 ml). The mixture was extracted with water (2 X 10 ml), dried (Na2SO4), and filtered to give a clear solution. The solution was added on to a column carrying strongly acidic cation exchange resin (0.3 mmol/g resin), which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 22 mg; 13C-NMR (CD3OD5 rotamers): δ 19.9, 26.3, 27.7, 39.9, 40.0, 43.6, 43.9, 44.4, 51.5, 53.6, 126.1, 126.7, 128.2, 128.4, 128.6, 128.9, 129.4, 129.6, 130.7, 130.9, 131.2, 131.6, 132.5, 132.7, 133.9, 134.1, 134.4, 134.8, 135.9, 136.3, 137.2, 172.4, 172.9.
Example 39 - N-ff4-MemylphenyIWmviyN-ri-fphenylmewylWrrolidin-3-yl>N'- phenylmet hylcarbamide f26HCH50)
[0237 J To a solution of 3-amino-l-phenylmethylpyrrolidine (353 mg, 2 mmol) and 4-methylbenzaldehyde (361 mg, 3 mmol) in methanol (20 ml) was added acetic acid in methanol (2 M, 6.7 ml) followed by NaCNBH3 in methanol (0.3 M, 3 ml). The mixture was stirred at room temperature. After 24 h, water (5 ml) was added. The mixture was stirred for another hour before concentrated. Flash chromatography in dichloromethane/methanol 10/1 gave N-((4-methylphenyl)methyl)amirio-l- phenylmethylpyrrolidine.
[0238] N-((4-Methylphenyl)methyl)amino-l-phenylmethylpyrrolidϊne (35 mg, 0.125 mmol) was dissolved in dichloromethane (1.5 ml), and benzylisocyanate (0.09 ml, 0.3 mmol) was added. The reaction mixture was stirred at room temperature. After 48 h, the crude reaction mixture was added on to a column carrying strongly acidic cation exchange resin, which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated to give the title compound. Yield: 48 mg, 92%; 13C-NMR (CD3OD): δ 20.0, 29.7, 44.2, 51.3, 53.4, 56.4, 57.8, 58.7, 126.8, 127.1, 127.3, 127.6, 128.3, 128.4, 128.9, 129.1, 135.9, 136.8, 140.3, 158.5.
Example 40 - N-(r4-Methylphenvπmethyl)-N-(l-(phenylmethvπpyrrolidin-3-yl)-4- methoxyphe nylacetamide (26HCH52)
[0239] To a solution of 3-amino-l-phenylmethylpyrrolidine (353 mg, 2 mmol) and 4-methylbenzaldehyde (361 mg, 3 mmol) in methanol (20 ml) was added acetic acid in methanol (2 M, 6.7 ml) followed by NaCNBH3 in methanol (0.3 M, 3 ml). The mixture was stirred at room temperature. After 24 h, water (5 ml) was added. The mixture was stirred for another hour before concentrated. Flash chromatography in dichloromethane/methanol 10/1 gave N-((4-methylphenyl)methyl)amino-l- phenylmethylpyrrolidine.
[0240] To a solution of N-((4-Methylphenyl)methyl)amino-l- phenylmethylpyrrolidine (35 mg, 0.125 mmol), diisopropylethylamine (0.14 ml) in dichloromethane (1.5 ml) was added 4-methoxyphenylacetyl chloride (0.1 ml, 0.5 mmol). The reaction mixture was stirred at room temperature. After 48 h, the crude reaction mixture was concentrated and redissolved in methanol. The solution was added on to a column carrying strongly acidic cation exchange resin, which was washed with methanol (3 X 6 ml), and eluted with 10% NH3 in methanol, and concentrated. Flash chromatography in dichloromethane/methanol 10/1 gave the title compound. Yield: 20 mg, 38%; 13C-NMR (CD3OD): δ 21.3, 30.2, 40.8, 47.8, 53.6, 53.9, 55.5, 57.5, 60.2, 114.4, 125.7, 127.0, 127.1, 127.3, 127.4, 128.4, 128.5, 128.7, 128.9, 129.2, 129.8, 130.0, 135.9, 137.0, 158.6.
Example 41 - N-((4-Methylphenyl)methylVN-π-φhenylmethyl)piperidin-4-yl)-4- methoxyphen ylthioacetamide (RO)
[0241] A mixture of N-((4-Methylphenyl)methyl)-N-(l-
(phenylmethyl)piperidin-4-yl)-(4-methoxyph enylmethyl)acetamide (20 mg, 0.045 mmol) and Lawesson's reagent (25 mg, 0.062 mmol), was taken in a glass vial and mixed thoroughly with magnetic stirbars. The glass vial was then irradiated in a microwave oven (900 W5 Whirlpool M401)for 8 min. Upon completion of the reaction, the yellow-colored material was transferred to an ion-exchange column with the aid of methanol (2 ml). The ion-exchange column was subsequently washed with CH2CI2 (2ml) and methanol (2 ml) and the product was thereafter eluted from the ion-exchange column (10% NH3 in methanol, 2 ml)to give N-((4-Methylphenyl)methyl)-N-(l-(phenylmethyl)piperidin-4-yl)- 4-methoxyphe nylmethyl thioacetamide (20 mg, 97%) as a white solid; LC-MS: (N+H)+ 459, tr 9.60 min; TLC (CH2Cl2 /methanol 20: l)Rf=0.38.
Pharmalogical Data
Example 42 - Receptor Selection and Amplification (R-SAT) Assays.
[0242] The functional receptor assay, Receptor Selection and Amplification Technology (R-SAT), was used (with minor modifications from that previously described U.S. Pat. No. 5,707,798) to screen compounds for efficacy at the 5-HT2A receptor. Briefly, NIH3T3 cells were grown in 96 well tissue culture plates to 70-80% confluence. Cells were transfected for 12-16 hours with plasmid DNAs using superfect (Qiagen Inc.) as per manufacture's protocols. R-SAT's were generally performed with 50 ng/well of receptor and 20 ng/well of Beta-galactosidase plasmid DNA. All receptor and G-protein constructs used were in the pSI mammalian expression vector (Promega Inc) as described in U.S. Pat. No. 5,707,798. The 5HT2A receptor gene was amplified by nested PCR from brain cDNA using the oligodeoxynucleotides based on the published sequence (see Saltzman et. al. Biochem. Biophys. Res. Comm. 181 :1469-78 (1991)). Large-scale transfections, cells were transfected for 12-16 hours, then trypsinized and frozen in DMSO. Frozen cells were later thawed, plated at 10,000-40,000 cells per well of a 96 well plate that contained drug. With both methods, cells were then grown in a humidified atmosphere with 5% ambient CO2 for five days. Media was then removed from the plates and marker gene activity was measured by the addition of the beta-galactosidase substrate ONPG (in PBS with 5% NP-40). The resulting colorimetric reaction was measured in a spectrophotometric plate reader (Titertek Inc.)at 420 nM. All data were analyzed using the computer program XLFit (IDBSm). Efficacy is the percent maximal repression compared to repression by a control compound (ritanserin in the case of 5HT2A). pIC50 is the negative of the log(IC50), where IC50 is the calculated concentration in Molar that produces 50% maximal repression. The results obtained for six compounds of the invention are presented in Table 1.
Table 1. Efficacy of Compounds at the 5-HT2A Receptor Efficacy.
Figure imgf000101_0001
Example 43 - In Vitro Efficacv of 26HCH17 as an Inverse Aeonist at the 5-HT2A
Receotor.
[0243] The graph shown in Figure 1 represents the data obtained from a dose response analysis of 26HCH17 and ritanserin as 5-HT2A receptor inverse agonists. Briefly, the 5-HT2A receptor, and the alpha subunit of the guanine nucleotide binding protein Gq were transiently transfected into NIH3T3 cells and assayed using the functional receptor assay, Receptor Selection and Amplification Technology (R-SAT) essentially as disclosed in U.S. Pat. No. 5,707,798. Each compound was screened at seven serially diluted concentrations in triplicate. Data were analyzed using least squares fit analysis with GraphPad Prism (San Diego, Calif.), and are reported normalized to percent response.
Example 44 - Selectivity Profile of Inverse Agonist 26HCH16D [0244] R-SAT assays (as described in Example 42) were carried out with cells transfected with receptors (listed below) to determine the receptor selectivity profile for compound 26HCH16D. 5HT2A inverse agonist data (IC50 nM; % efficacy) were derived from detailed dose response curves (7 points in triplicate). All other data (initial concentration at which at least 30% efficacy observed; actual efficacy figure) derived from the 4 dose profiling protocol in which compounds were tested at 4 doses in duplicate. nr=activity less than 30% at all doses tested (3, 30, 300, 3000 nM), therefore EC50/IC50 greater than 3000 nM). The results are presented in Table 2.
Table 2. Profile of 5-HT2A Inverse Agonist 26HCH16D.
Figure imgf000102_0001
[0245] As indicated above, 26HCH16D is a highly selective 5-HT2A inverse agonist. General LC-MS Procedure for Working Examples ELHO1-46. MBT01-14 and AKUOl- 38
[0246] In the following examples, HPLC/MS analyses were performed using either of two general methods (Method A or Method B). The tr values reported below were obtained using one of these procedures, as indicated in the specific examples.
[0247] The methods were as follows: Method A: Agilent HPl 100 HPLC/MSD.
[0248] G1312A Binary pump, G1313A Autosampler, G1316A Column compartment, G1315A Diode array detector (190-450 nm), 1946 A MSD, electrospray ionization.
[0249] Chromatography: 8 mM ammoniumacetate in water/acetonitrile.
[0250] Gradient start at 70% org. up to 100% org. over 12 min, down to 70% org. over 0.5 min, held for 3.5 min. Total runtime 16 min. Flowrate 1 ml/min
[0251] Column, Phenomenex Luna Cl 8 (2) 3 um 75 X 4.6 mm.
[0252] MS Parameters: Drying gas, 10 1/min. Nebulizer pressure, 40 psig. Gas temp, 350 C. VCap, 4000. Method B: Waters/Micromass HPLC/MS
[0253] 600 LC-pump, 2700 Sample manager, 2487 Dual absorbance detector (channel A-205 nm, channel B-235 nm), Micromass ZMD-mass-spectrometer, electrospray ionization.
[0254] Chromatography: 0.15% TFA in water/acetonitrile.
[0255] Gradient start at 30% org. up to 100% org. over 10 min, held for 3 min. down to 30% org. over 0.5 min, held for 4.5 min. Total run time 18 min. Flowrate, 1 ml/min.
[0256] Column, Symmetry C 18, 5μm, 4.6 X 50 mm. or 10 mM ammoniumacetate in water/acetonitrile.
[0257] Gradient start at 30% org. for 2.5 min, up to 100% org. over 10 min, held for 9 min, down to 30% org. over 0.5 min, held for 5 min. Total run time 27 min. Flowrate, 1 ml/min.
[0258] Column: Phenomenex Synergi C 12, 4μm, 4.6 X 50 mm. [0259] MS Parameters: Desolvation Gas, 404 1/H. Capillary, 5.3 kV. Cone, 36V. Extractor, 3V. Source Block Temp, 130 C. Desolvation Temp, 250 C.
Example 45 - 2-r4-methoxyphenylVN-(4-methylbenzylVN-('piperidin-4-yl>acetamide (50ELHS?^
[0260] Reaction Step 1 : N-trifluoroacetyl-4-piperidone (50ELH84)
[0261] 4-Piperidone hydrochloride monohydrate (4.0 g, 26 mmol, 1.0 eq) was dissolved in 130 ml of dichloromethane. After addition of triethylamine (8.66 g, 3.3 eq) the reaction mixture was stirred for 10 min. The mixture was cooled on an ice-bath (0° C). Trifluoroacetic anhydride (12.0 g, 2.2 eq) was added dropwise under stirring. After 2 hours the reaction was quenched by addition of distilled water. The aqueous phase was extracted twice with dichloromethane. The combined organic layers were collected and dried with sodium sulfate. Concentration afforded N-trifluoroacetyl-4-piperidone.
[0262] Reaction Step 2: 4-(4-Methylben2ylamino)-l-(trifluoroacetyl)piperidin (50ELH85)
[0263] Methanol (150 ml) was added to an Erlenmeyer flask and acetic acid was added under stirring until pH 5.4-Methylben2ylamine (3.14 g, 25.9 mmol) and N- trifluoroacetyl-4-piperidone(from reaction step 1) (5.065 g, 25.9 mmol) were added to a 250 ml round-bottomed flask and dissolved in the methanol/acetic acid (150 ml) solution previously made. The reaction mixture was stirred for 5 min and NaCNBH3 (2.46 g, 38.9 mmol) was added slowly under stirring. After 20 hours the reaction was concentrated and transferred to a separatory funnel containing dichloromethane and distilled water. The aqueous phase was made basic by addition OfNa2CO3. The aqueous phase was extracted twice with dichloromethane. The combined organic layers were collected and dried with Na2SO4. Concentration afforded, 4-(4-methylbenzylamine)-l-(trifluoroacetyl)piperidine. UV/MS 60/53 (M+ 301), tr (A, MS) 3.267.
[0264] Reaction Step 3: 2-(4-Methoxyρhenyl)-N-(4-methylbenzyl)-N-(l- trifluoroacetylpiperidin-4-yl) acetamide (50ELH86)
[0265] The product from reaction step 2 (7.8 g, 25.9 mmol) was dissolved in 100 ml of dichloromethane and stirred while 4-methoxyphenylacetyl chloride (4.8 g, 25.9 mmol) was added. After 4 hours, heptane was added whereupon the product precipitated as the hydrochloride salt. The solvent was removed by evaporation. The crude material was purified by flash chromatography EtOAc/Heptane (1 :2) Yield (overall: Reaction steps 1+2+3) 3.912 g (34% ), UV/MS 91/58 (M+ 449), tr (A3 MS) 4.319. 1H-NMR (400 MHz, CDCl3) δ 6.80-7.15 (Ar, 4H)3 4.64 (brt, IH), 4.4 (s, 2H), 3.95 (d, 2H), 3.72 (s, 3H)5 3.50 (s, 2H)5 3.09 (t, 2H), 2.7 (t5 2H), 2.32 (s, 3H)5 1.75 (btr, 2H). 13C-NMR δ 172.5; 158.8; 137.4; 134.9; 129.9; 129.9; 129.8; 127.1; 125.8; 114.3; 55.4; 52.2; 47.3; 45.3; 43.4.40.6; 30.1; 29.2; 21.2.
[0266] Reaction Step 4: 2-(4-Methoxyphenyl)-N-(4-methylbenzyl)-N- (piperidin-4-yl) Acetamide (50ELH87)
[0267] The product from reaction step 3 (3.9 g, 8.7 mmol) was dissolved in methanol (12 ml). In a 250 ml round bottom flask a saturated solution of potassium carbonate in methanol was prepared. To this solution, the 2-(4-methoxyphenyl)-N-(4- methylbenzyl)-N-(N-trifluoroacetpiperidin-4-yl)ac etamide solution was added under stirring. After 4 hours, the solution was concentrated and the remaining solid taken upin base and dichloromethane. The combined organic layers were dried with sodium sulfate and concentrated. UV/MS 91/72 (M+ 353), tr (A, MS) 2.210.
[0268] The corresponding hydrochloride salt was also prepared, by dissolving the free base in dichloromethane (1 ml) and HCl (1 eq. 2 M HCl in ether) was added with stirring. The salt was precipitated by addition of the dichloromethane solution into heptane. Concentration on the rotary evaporator returned the product is white crystals.
Example 46 - 2-r4-MethoxyphenylVN-f4-methylbenzyl')-N-(l-methylpiperidin-4-yl) acetamide f50ELH27>
[0269] Reaction Step 1 : 4-(4-Methylbenzylamino)-l-methylpiperidine (50ELH25)
[0270] Methanol (50 ml) was added to an Erlenmeyer flask and acetic acid was added under stirring until pH 5. Methylbenzylamine (1.0 g, 8.8 mmol) and 1-Methyl- 4-piperidone (1.1 g, 8.8 mmol) were added to a 100 ml round-bottomed flask and dissolved in the methanol/acetic acid (40 ml) solution previously made. The reaction mixture was stirred for 5 min and NaCNBH3 (0.83 g, 13.2 mmol) was added slowly under stirring. After 20 hours the reaction was concentrated and transferred to a separatory funnel containing dichloromethane and distilled water. The aqueous phase was made basic by addition of Na2CO3. The aqueous phase was extracted twice with dichloromethane. The combined organic layers were collected and dried with Na2SO4. Concentration afforded the title compound. Yield (crude): 98%. UV/MS 89/88.(M+ 353), tr (A, MS) 3.982.
[0271] Reaction Step 2: 2-(4-Methoxyphenyl)-N-(4-methylbenzyl)-N-(l- methylpiperidin-4-yl) acetamide (50ELH27)
[0272] The product from reaction step 1 (1.9 g, 8.7 mmol) was dissolved in 40 ml of dichloromethane and stirred while 4-methoxyphenylacetylchloride (1.606 g, 8.7 mmol) was added. After 4 hours, heptane was added whereupon the product precipitated as the hydrochloride salt. The solvent was removed by evaporation. The crude material was purified by flash chromatography first eluting with 10% MeOH in CH2CI2 and thereafter eluting with 0-20% MeOH in CH2Cl2 and 5% NEt3. Yield (overall: Reaction steps 1+2): 77%. UV/MS: 100/100 (M+ 367), tr (A, MS) 4.359, Rf 0.15 (2% MeOH in CH2Cl2). 1H-NMR (400 MHz, CDCl3) δ 12.6 (s, IH), 7.16 (d, J=7.0 Hz5 2H), 7.10 (d, J=7.0 Hz, 2H), 7.04 (d, J=8.0 Hz5 2H)5 6.82 (d, J=8.0 Hz5 2H)5 4.87 (tt, J=I LO5 4.0 Hz, IH)5 4.53 ppm (s, 2H)5 3.78 (s5 3H), 3.55 (s, 2H), 3.42 (brd, J=I LO Hz5 2H)5 2.80 (brq, J=I LO Hz5 2H), 2.7 (d, J4.0 Hz, 3H), 2.42 (dq, J=13.0, 3.0 Hz, 2H), 2.34 (s, 3H), 1.78 (brd, J=13.0 Hz, 2H). 13C-NMR δ 173.1; 158.9; 137.4; 134.8; 129.9; 126.7; 125.8; 114.4; 76.9; 55.5; 54.6; 48.8; 43.7; 40.5; 26.4; 21.2
Example 47 - 2-(4-Methoxyphenyl)-N-(4-methylbenzyl')-N-π-cvclohexylmethylpiperidin- 4-Vl) acetamide (42ELH45)
[0273] 50ELH87 (the hydrochloride salt) (0.5 g, 1.29 mmol, LO eq) was dissolved in ethanol (100 ml). Cyclohexanecarboxaldehyde (2.5 g, 20 eq.) was added followed by addition of sodium borohydride (0.084 g, 2.0 eq.). The reaction was stirred for 36 h and acetic acid (3 ml) was added. The reaction was stirred for additionally 2 h and extracted with sodium hydrogen carbonate (3 times) and dichloromethane. The organic layers were dried with sodium sulfate and concentrated. The product was purified by flash chromatography (1-10% MeOH in CH2CI2). The resulting product was dissolved in ether (20. ml) and MeOH (added dropwise until dissolved) and HCl (1 eq. 2 M HCl in ether) was added under stirring. The hydrochloride salt of 2-(4~methoxyphenyl)-N-(4- methylbenzyl)-N-(l-cyclohexylmethylpiperidin-4-yl ) acetamide precipitated and the white crystals were filtered. Yield 80 mg (16%), UV/MS 100/100 (M+ 449), rt (A, MS) 7.105, mp 133-135° C5 Rf0.25 (2% MeOH/CH2Cl2). 1H-NMR (400 MHz, CDCl3) δ 11.9 (brs, IH), 7.12 (q, 4H), 7.02 (d, 2H), 6.80 (d, 2H), 4.87 (m, IH), 4.58 (s, 2H), 3.77 (s, 3H)5 3.55 (s, 2H), 3.48 (m, 2H), 2.70 (m, 4H), 2.31 (s, 3H), 1.91 (d, 2H), 1.75 (m, 3H)5 1.64 (d5 IH), 1.22 (d, 2H)5 1.13 (tt, 2H), 1.02 (brq, 2H). 13C-NMR δ 173.1; 158.8; 137.2; 135.1; 129.9; 129.8; 126.8; 125.8; 114.4; 64.1; 55.5; 53.4; 49.2; 46.5; 40.4; 33.9; 25.9; 25.8; 25.7; 21.2.
Example 48 - 2-r4-Methoxyphenyl*)-N-(r4-methylbenzyl)-N-π-ethylpiperidin-4- yliacetamide C42ELH80>)
[0274] 50ELH87 (0.25 g, 0.71 mmol, 1.0 eq) was dissolved in acetonitrile (15 ml) and ethyl bromide (0.232 g, 3.0 eq.) was added under stirring. After 2 min Hunings base (0.084 g, 10.0 eq.) was added. After 36 hours, the solution was extracted with sodium hydrogen carbonate solution and dichloromethane (3 times). The organic layers were dried with sodium sulfate and concentratedyielding a yellow oil. The product was purified by flash chromatography (2% MeOH in CH2Cb). The resulting product was dissolved in dichloromethane (1 ml) and HCl (1 eq. 2 M HCl in ether) was added under stirring. The salt was precipitated by addition of the dichloromethane solution into heptane. Concentrationon the rotary evaporator gave the product as white crystals. Yield 170 mg (63%), UV/MS 98/95 (M+ 381), mp 153-155° C, rt (A, MS) 3.033, Rf0.35 (3% MeOH/CH2Cl2). 1H-NMR (400 MHz, CDCl3) δ 12.2 (s, IH)3 7.15 (d, 2H)5 7.12 (d5 2H)5 7.08 (d, 2H), 6.82 (d, 2H), 4.89 (m, IH), 4.58 (s, 2H), 3.79 (s, 3H), 3.58 (s, 2H), 3.50 (d, 2H), 2.90 (m, IH), 2.7 (brq, 2H), 2.45 (m, 2H), 2.34 (s, 3H), 1.80 (d,, 2H), 1.44 (t, 3H). 13C-NMR δ 173.1; 158.9; 137.3; 134.9; 129.9; 125.8; 114.4; 55.5; 52.3; 52.0; 49.2; 46.5; 40.5; 26.2; 21.2; 9.5.
Example 49 - 2-f4-Methoxyphenyl)-N-(4-chlorbenzviyN-π-ethylpiperidin-4-yl) acetamide f42ELH85\
[0275] This compound was prepared similarly to 50ELH27
[0276] Reaction-Step 1: (42ELH84)
[0277] Starting materials: l-Methyl-4-piperidone (0.5 g, 4.4 mmol,- 1.0 eq.), 4- chlorobenzylamine (0.626 g, 1.0 eq.), sodium cyanoborohydride (0.279 g5 1.5 eq.).
[0278] Reaction-Step 2: (42ELH85)
[0279] Starting materials: 42ELH84, 4-methoxyphenylacetylchloride (0.774 g, 1.0 eq.). [0280) The procedure was analogous to 50ELH27, but the product was purified by ion-exchange chromatography followed by HPLC. The hydrochloride salt was made by dissolving the free base in dichloromethane (1 ml) and HCl (1 eq. 2 M HCl in ether) was added under stirring. The salt was . precipitated by addition of the dichloromethane solution into heptane followed by concentration on the rotary evaporator.
[0281] Product White crystals. UV/MS 98/97 (M+ 387), rt (A, MS) 2.953. 1H- NMR (400 MHz5 CDCl3) δ 12.6 (s, IH), 7.35 (d, 2H)3 7.18 (d, 2H), 7.05 (d, 2H), 6.82 (d, 2H), 4.89 (m, IH), 4.55 (s, 2H), 3.80 (s, 3H), 3.55 (s, 2H), 3.45 (brs, 2H), 2.80 (brs, 2H), 2.72 (s, 3H), 2.25 (brs, 3H)5 1.80 (brs, 2H). 13C-NMR δ 173.0; 158.9; 136.5; 133.6; 129.8; 129.4; 127.3; 126.3; 114.5; 55.5; 54.6; 48.7; 46.3; 43.7; 40.5; 26.3.
Example 50 - 2-(4-MethoxyphenyI)-N-(4-methylbenzyQ-N-('l-isopropylpiperidin-4- vDacetamide f42ELH79).
. [0282] Procedure as 42ELH80
[0283] Starting materials: 50ELH87 (0.25 g, 0.71 mmol, 1.0 eq.), isopropylbromide (0.262 g, 3.0 eq.).
[0284] Product: Yield 130 mg (46%), UV/MS 100/100 (M+395), rt (A, MS) 3.360. 1H-NMR (400 MHz, CDCl3) δ 12.0 (s, IH), 7.15 (d, 2H), 7.10 (d, 2H), 7.05 (d, 2H), 6.82 (d, 2H), 4.87 (m, IH), 4.60 (s, 2H), 3.79 (s, 3H), 3.57 (s, 2H), 3.38 (brd, 3H), 2.79 (q, 2H), 2.63 (q, 2H), 2.34 (s, 3H), 1.80 (d, 2H), 1.39 (d, 6H). 13C-NMR δ 173.1 ; 158.9; 137.3; 135.1; 129.8; 126.8; 125.8; 114.4; 57.9; 49.4; 48.2; 46.5; 40.5; 25.9; 21.2; 16.9.
Example 51 - 2-('4-Methoxyphenyl')-N-(4-chlorobenzyl')-N-(piperidin-4-vπacetamide (42ELH89) ( As Starting Material in Other Reactions. Used Unpurified)
[0285] Procedure as 50ELH27.
[0286] Reaction Step 1 : N-Trifiuoroacetyl-4-piperidone (42ELH86)
[0287] Starting materials: 4-Piperidone hydrochloride monohydrate (2.0 g, 13 mmol, 1.0 eq), trifluoroacetic anhydride (6.0, 2.2 eq.). TLC showed full conversion.
[0288] Product: Rf 0.9 (10% MeOH/CH2Cl2). [0289] Reaction Step 2: 4-(4-Chlorobenzoylamino)~l-
(trifluoroacetyl)piperidin (42ELH87)
10290] Starting materials: 42ELH86 (2.5 g, 12.8 mmol, 1.0 eq.), 4- Chlorobenzylamine (1.8 g, 1.0 eq.)
[0291] Reaction Step 3: 2-(4-Methoxyphenyl)-N-(4-chlorobenzyl)-N-(l- trifluoroacetylpiperidin-4-yl) acetamide (42ELH88)
[0292] Starting materials: 42ELH87 (4.0 g, 12.5 mmol. 1.0 eq.), 4- methoxyphenylacetylchloride (2.31 g, 1.0 eq.)
[0293] Reaction Step 4: 2-(4-Methoxyphenyl)-N-(4-chlorobenzyl)-N- (piperidin-4-yl)acetamide (42ELH89)
[0294] Product: Yield: 2 g (57%), UV/MS 80/82 (M+ 373), Rf 0.2 (50% EtOAc/Heptane).
Example 52 - 2-f4-MemoxyphenvD-N-(4-chlorbenzyl)-N-π-cvclopentylpiperidin-4- vDacetam ide (42ELH9U.
[0295] Procedure as 42ELH80, but the product was purified by HPLC. The acidic eluent was made basic with sodium carbonate and extracted with dichloromethane (3 times). The combined organic layers were collected and dried with sodium sulfate and concentrated. The remaining product was dissolved in 1 ml of dichloromethane and HCl (1 eq. 2 M HCl in ether) was added under stirring. This solution was added drop- wise to a large excess of n-heptane to make the hydrochloride precipitate. The solvent was evaporated off to form white crystals of 2-(4-methoxyphenyl)-N-(4-chlorbenzyl)-N-(l- cyclopentylpiperidin-4-yl)aceta mide, hydrochloride.
[0296] Starting materials: 42ELH89 (0.25 g, 0.67 mmol, 1.0 eq.), cyclopentylbromide (0.3, 3.0 eq.)
[0297] Product: Yield. 211.2 mg (76%). Purification by ion-exchange: UV/MS 90/98. Purification by HPLC UV/MS 100/100 (M+ 441), Rf 0.2 (3% MeOH/CH2Cl2), r, (A, MS) 4.067. 1H-NMR (400 MHz, CDCl3) 512.2 (brs, IH)3 7.32 (d, 2H), 7.17 (d, 2H), 7.04 (d, 2H), 6.82 (d, 2H), 4.90 (brt, IH), 4.58 (s, 2H), 3.79 (s, 3H), 3.58 (brd, 2H), 3.54 (s, 2H)3 3.14 (brq, 2H), 2.58 (brq, 2H)5 2.04 (m, 4H)5 1.89 (m, 4H), 1.75 (brd, 2H). 13C-NMR δ 173.0; 158.9; 133.5; 129.8; 129.3; 127.3; 126.4; 114.5; 68.4; 55.5; 51.9; 49.1; 46.2; 40.5; 28.5; 26.0; 23.8. Example 53 - 2-(4-MethoxyphenylVN-('4-chlorbenzylVN-('l-isopropylpiperidin-4- vDacetamid e (42ELH90V
|0298] 42ELH89 (0.25 g, 0.67 mmol, 1.0 eq) was transferred to a 4 ml vial and dissolved in acetonitrile (2 ml). Isopropyl bromide (0.25 g, 3.0 eq.) was added along with Hunigs base (0.87 g, 10.0 eq.). The vial was sealed and shaken for 4 days at 60° C. The reaction mixture was transferred to a separatory funnel with distilled water and CH2CI2. The aqueous phase was made basic with sodium hydrogen carbonate and extracted with dichloromethane (3 times). The organic layers were collected and dried with sodium sulfate and concentrated, this resulted in a yellow oil. The product was purified by flash chromatography (3% MeOH in CH2Ck). The resulting product was dissolved in dichloromethane (1 ml) and HCl (1 eq. 2 M HCl in ether) was added under stirring. The salt precipitated by addition of the dichloromethane solution into heptane. Concentration on the rotary evaporator returned the product as white crystals. Yield 101.2 mg (63%), UV/MS 94/96 (M+ 415), Rf0.25 (3% MeOH/CH2Cl2).
102991 1H-NMR (400 MHz, CDCl3) δ 12.05 (brs, IH), 7.36 (d, 2H), 7.18 (d,
2H), 7.04 (d, 2H), 6.82 (d, 2H), 4.88 (rn, IH), 4.60 (s, 2H)5 3.79 (s, 3H), 3.55 (d, 2H), 3.36 (d, 3H), 2.80 (brq, 2H), 2.65 (brq, 2H), 1.76 (brd, 2H), 1.39 (d, 6H). 13C-NMR δ 173.0; 159.0; 137.0; 136.0; 129.7; 129.3; 127.4; 126.4; 114.5; 57.9; 55.5; 49.2; 48.2; 46.2; 40.5; 25.8; 16.9.
Example 54 - 2-(PhenylVN-(4-trifluoromethylbenzyl)-N-(l-methylpiperidin-4- vDacetamide (50ELHUb)
[0300] Procedure as for 50ELH27. Purification was done by HPLC. The hydrochloride salt was made by dissolving the free-base in dichloromethane (1 ml) and HCl (1 eq. 2 M HCl in ether) was added under stirring. The salt was precipitated by addition of the dichloromethane solution into heptanefollowed by concentration.
[0301] Reaction-Step 1: 4-(4-Trifluoromethylbenzylamino)-l-methylρiperidin (50ELH2).
[0302] Starting materials: l-Methyl-4-piperidone (1.13 g, 10.0 mmol, 1.0 eq.), 4-trifluoromethylbenzyIamine (1.75 g, 1.0 eq.).
[0303] Product: UV/MS 80/92 (M+273). [0304] Reaction-Step 2: 2-(Phenyl)-N-(4-trifluoromethylbenzyl)-N-(l- methylρiperidin-4-yl)acetamide (50ELH14b).
[0305] Starting materials: 50ELH2 (0.12 g, 0.44 mmol, 1.0 eq.), phenylacetylchloride (0.068 g, 1.0 eq.).
[0306] Product: UV/MS 100/97 M+ 390), rt (A, MS) 3.797, Rf 0.3 (5% MeOH/CH2Cl2). 1H-NMR (400 MHz, CDCl3, rotamers 54/46) δ 7.52 (d, 2H), 7.42 (d, 2H), 7.12 7.30 (m, 4H5), 4.63 and 3.74 (2m, IH), 4.38 (brs, 2H), 3.80 and 3.50 (2s, 3H), 3.31 and 2.78 (2d, 2H), 2.33 and 2.18 (2s, 2H), 2.24 and 1.65-1.90 (t and m, 4H), 1.60 and 1.22 (2d, 2H), 1. 13C-NMR δ 172.3; 171.8; 143.9; 135.1; 134.8; 129.1; 129.0; 128.9; 128.7; 127.4; 127.3; 127.2; 126.3; 126.1; 126.0; 56.0; 55.2; 54.9; 50.9; 46.8; 45.2; 44.9; 42.2; 41.7; 30.6; 28.4.
Example 55 - 2-(4-Fluorophenvπ-N-(4-trifluoromethylbenzyl)-N-(l-methylpiperidin-4- vDac etamide (50ELHHc*)
[0307] Procedure as 50ELH14B.
[0308] Reaction-Step 2: 2-(4-Fluorophenyl)-N-(4-trifluoromethylbenzyl)-N- (l-methylpiperidin-4-yl)a cetamide (50ELH14c).
[0309] Starting materials: 50ELH2 (0.12 g, 0.44 mmol, 1.0 eq.), 4- fluorophenylacetylchloride (0.076 g, 1.0 eq.).
[0310] Product: Yield 69.7 mg (36%), UV/MS 100/98 (M+ 409), rt (A, MS) 3.839, Rf 0.3 (5% MeOH/CH2Cl2). 1H-NMR (400 MHz, DMSO, rotamers 65/35) δ 10.80 and 10.60 (2s, IH), 7.71 and 7.62 (2d, 2H), 7.47 and 7.38 (2d, 2H), 7.00-7.36 (t and m, 4H), 4.70 and 4.50 (2s, 2H), 4.30 (m, IH), 3.93 and 3.56 (2s, 2H), 3.34 (s, 2H), 3.00 (brq, 2H), 2.64 (s, 3H), 2.08 (m, 2H), 1.68 and 1.58 (2d, 2H). 13C-NMR δ 176.8; 176.4; 167.6; 165.3; 150.0; 149.0; 136.6; 132.5; 131.0; 130.5; 120.6; 120.5; 120.5; 120.4; 58.1; 58.0; 57.0; 54.5; 52.0; 49.3; 47.6; 45.0; 32.4; 31.4.
Example 56 - 2-r4-Methoxyphenyl)-N-(r4-trifluoromethvIbenzvπ-N-π-methylpiperidin-4- vOa cetamide (50ELHUd)
[0311] Procedure as 50ELH14B.
[0312J Reaction-Step 2: 2-(4-Methoxyphenyl)-N-(4-trifluoromethyIbenzyl)-N- (l-methylpiperidin-4-yl) acetamide (50ELH14d). [0313] Starting materials: 50ELH2 (0.15 g, 0.55 mmol, 1.0 eq.), A- methoxypheπylacetylchloride (0.1 g, 1.0 eq.).
[0314] Product: Yield 57.5 mg (29%), UV/MS 99/100 (M+ 421), r, (B, MS) 6.30, Rf0.25 (3% MeOH/CH2Cl2). 1H-NMR (400 MHz, CDCl3) δ 12.4 (brs, IH)5 7.55 (d, 2H), 7.28 (d, 2H), 6.96 (d, 2H), 4.84 (brt, IH), 4.59 (s, 2H), 3.72 (s, 3H), 3.46 (s, 2H), 3.38 (d, 2H), 2.78 (q, 2H), 2.64 (s, 3H)5 2.38 (q, 2H), 1.70 (d, 2H). 13C-NMR δ 173.0; 159.0; 142.3; 130.0; 129.8; 126.3; 126.2; 114.7; 114.5; 55.5; 54.4; 48.7; 46.5; 43.6; 40.6; 26.3.
Example 57 - 2-r4-TrifluoromethylphenylVN-(4-trifluoromethylbenzyl)-N-(l- methylpiperidi n-4-γl")acetamide (5 OELH 14a)
[0315] Procedure as 50ELH14B.
[0316] Reaction-Step 2: 2-(4-Trifluoromethylphenyl)-N-(4- trifluoromethylbenzyl)-N-(l-methylpiperid in-4-yl)acetamide (50ELH14a).
[0317] Starting materials: 50ELH2 (0.12 g, 0.44 mmol, 1.0 eq.), A- trifluoromethylphenylacetylchloride (0.1 g, 1.0 eq.).
[0318] Product: Yield 92.6 mg (42%), UV/MS 89/93 (M+ 458), rt (A, MS) 4.211, Rf0.3 (5% MeOH/CH2Cl2). 1H-NMR (400 MHz, CDCl3) δ 12.7 (brs, IH)5 7.56 (d, 2H), 7.48 (d, 2H)5 7.17 (d, 2H), 4.86 (m, IH), 4.63 (s, 2H), 3.58 (s, 3H), 3.40 (d, 2H), 2.75 (q, 2H)3 2.65 (d, 3H), 2.46 (dq, 2H)5 1.73 (brs, 2H). 13C-NMR δ 171.8; 141.9; 138.4; 129.4; 127.9; 126.3; 126.3; 126.2; 125.9; 125.8; 54.4; 48.8; 46.6; 43.6; 40.9; 26.2.
Example 58 - 2-(4-Fluorophenyl)-N-C4-fluorobenzviyN-π-methylpiperidin-4- vftacetamide f50ELH6)Procedure as 50ELH14B.
[0319] Reaction-Step 1: 4-(4-Fluorobenzylamino)-l-methylpiperidine (50ELH4).
[0320] Starting materials: l-Methyl-4-piperidone (1.13 g, 10.0 mmol, 1.0 eq.), 4-fluorobenzylamine (1.25 g, 1.0 eq.).
[0321] Product: Yield 2.154 g (97%), UV/MS 79/89 (M+ 223).
[0322] Reaction-Step 2: 2-(4-Fluorophenyl)-N-(4-fluorobenzyl)-N-(l- methylpiperidin-4-yl)acetamide (50ELH14a).
-I l l- [0323] Starting materials: 50ELH4 (0.12 g, 0.54 mmol, 1.0 eq.), 4- fluorophenylacetylchloride (0.096 g5 1.0 eq.).
[0324] Product: Yield 57 mg (29%), UV/MS 100/100 (M+ 359), r, (A, MS) 3.763, Rf0.25 (3% MeOH/CH2Cl2). 1H-NMR (400 MHz, CDCl3) 67 12.6 (brs, HH)5 7.2 (dd, 2H), 7.06 (m, 4H), 6.98 (t, 2H), 4.88 (tt, IH), 4.58 (s, 4H), 3.45 (d, 2H), 2.81 (q, 2H), 2.72 (d, 3H), 2.48 (brq, 2H), 1.78 (brs, 2H). 13C-NMR δ 172.5; 163.4; 160.8; 133.4; 130.6; 130.2; 127.5; 127.4; 116.3; 116.1; 115.9; 115.7; 54.5; 48.8; 46.2; 43.6; 40.3; 26.3.
Example 59 - 2-(/4-Methoxyphenyl)-N-('4-fluorobenzyl')-N-ri-methylpiperidin-4- vnacetamide f50ELH8y
[0325] Procedure as 50ELH14B
[0326] Reaction-Step 2:
[0327] Starting materials: 50ELH4 (0.12 g, 0.54 mmol, 1.0 eq.), 4- methoxyphenylacetylchloride (0.1 g, 1.0 eq.).
[0328] Product: Yield 54 g (26%), UV/MS 100/100 (M+ 371), r, (A, MS) 3.257, Rf0.25 (3% MeOH/CH2Cl2). 1H-NMR (400 MHz, CDCl3) 67 12.2 (brs, IH), 7.12 (m, 2H), 6.97 (m, 4H)5 6.75 (d, 2H), 4.80 (bit, IH), 4.49 (s, 2H), 3.71 (s, 3H), 3.47 (s, 2H), 3.37 (d, 2H), 2.8 (q, 2H), 2.64 (s, 3H), 2.35 (q, 2H), 1.69 (d, 2H). 13C-NMR δ 173.0; 163.5; 161.1; 158.9; 133.7; 133.6; 129.8; 127.6; 127.5; 126.5; 116.2; 116.0; 114.6; 114.5; 55.5; 54.4; 48.8; 46.2; 43.6; 40.5; 26.4.
Example 60 - 2-(PhenylVN-r4-fluoroben2yl')-N-(l-methylpiperidin-4-yl*)acetamide CSOELHlO^
[0329] Procedure as 50ELH14B.
[0330] Reaction-Step 2: 2-(Phenyl)-N-(4-fluorobenzyl)-N-(l-methylpiperidin- 4-yl)acetamide (50ELH10).
[0331] Starting materials: 50ELH4 (0.13 g, 0.59 mmol, 1.0 eq.), phenylacetylchloride (0.091 g, 1.0 eq.).
[0332] Product: UV/MS 100/94 (M+ 341), r, (A, MS) 3.127, Rf 0.25 (3% MeOH/CH2Cl2). 1H-NMR (400 MHz, DMSO, rotamers 54/56) δ 12.38 (brs, IH), 7.35- 7.00 (m, 9H), 4.55 and 4.40 (2s, 2H), 4.50 and 4.25 (bit, IH), 3.91 and 3.56 (2s, 2H), 3.30 (Hidden under water signal) (2H), 2.98 (d, 2H), 2.64 (s, 3H), 2.09 (bit, 2H), 1.66 and 1.45 (2brd, 2H). 13C-NMR δ 171.9; 171.6; 162.8; 160.4; 136.5; 136.2; 135.4; 129.9; 129.7; 129.5; 129.2; 129.0; 128.9; 128.7; 127.2; 127.1; 116.2; 116.0; 115.6; 53.2; 52.5; 49.8; 46.9; 44.0; 42.8; 40.9; 40.6; 40.4; 40.2; 40.0; 39.8; 39.6; 27.7; 26.6.
Example 61 - 2-r4-Trifluoromethylphenvπ-N-(4-fluorobenzyl')-N-('l-methylpiperidin-4- vPac etamide (50ELH12.sup.2'>
[0333] Procedure as 50ELH14B.
[0334] Reaction Step 0: 4-Trifluoromethyphenylacetyl chloride (50ELH12.sup.l)
[0335] 4-Trifluorophenylacetic acid (1.0 g) and thionyl chloride (15 ml) were refluxed for 1 h. The excess thionyl chloride was evaporated off. NMR showed complete conversion.
[0336] Reaction-Step 2: 2-(4-Trifluoromethlphenyl)-N-(4-fluorobenzyl)-N-(l- methylpiperidin-4-yl)ac etamide (50ELH1 22).
[0337] Starting materials: 50ELH4 (0.12 g, 0.55 mmol, 1.0 eq.), 4- trifluoromethylphenylacetylchloride (50ELH12.sup.l) (0.11 g, 0.5 mmol, 1.0 eq.).
[0338] Product: Yield 47.1 mg (24%), UV/MS 96/96 (M+ 409), rt (A3 MS) 4.566, Rf 0.25 (3% MeOH/CH2Cl2). 1H-NMR (400 MHz, CDCl3)O 7.52 (d, 2H), 7.22 (d, 2H), 7.17 (dd, 2H), 7.04 (t, 2H), 4.86 (brt, IH), 4.58 (s, 2H), 3.64 (s, 2H)3 3.45 (brd, 2H), 2.84 (brq, 2H)5 2.71 (d, 3H), 2.45 (brq, 2H), 1.77 (brd, 2H). 13C-NMR δ 171.8; 163.6; 161.2; 138.7; 133.3; 129.8; 129.5; 127.5; 127.4; 125.8; 125.7; 116.4; 116.2; 54.4; 48.9; 46.3; 43.6; 40.8; 26.3.
Example 62 - 4-f4-Methoxybenzylamino)-l-methylpiperidine (50ELH18).
[0339] Procedure as 50ELH27.
[0340] Starting materials: l-Methyl-4-piperidone (1.13 g, 10.0 mmol, 1.0 eq.), 4-methoxybenzyIamine (1.37 g, 1.0 eq.).
[0341] Product: UV/MS 95/95 (M+ 235), rt (A, MS) 3.509. 1H-NMR (400 MHz, CDCl3) δ 7.3-6.8 (m, 4H), 3.77 (s, 3H), 3.73 (s, 2H), 2.86 (m, 2H), 2.55 (m, IH), 2.30 (s, 3H), 2.1 (t, 2H), 1.96 (dd, 2H), 1.50 (m, 2H). Example 63 - 2-("4-Trifluoromethylphenyl')-N-r4-fniethoxycarbonyl')benzyl')-N-(l- methylpiperi din-4-vDacetamide (50ELH20A)
[03421 Procedure as 50ELH14B.
[0343] Reaction-Step 1: Methyl 4-(N-[l-methylpiperidine-4-yl]aminomethyl) benzpate (50ELH19).
[0344] Starting materials: l-Methyl-4-piperidone (1.13 g, 10.0 mmol, 1.0 eq.), methyl 4-(aminomethyl)benzoate hydrochloride (2.0 g, 1.0 eq.).
[03451 Product: UV/MS 81/88 (M+ 263), rt (A, MS) 3.060. 1H-NMR (400 MHz5 CDCl3) δ 8.00 (d, 2H). 7.20 (d, 2H), 3.90 (s, 3H), 3.85 (s, 2H), 2.96 (dt, 2H), 2.7 (brs, IH), 2.62 (m, IH), 2.40 (s, 3H)3 2.28 (t, 2H)3 1.96 (m, 2H), 1.56 (m, 2H).
[0346J Reaction-Step 2: 2-(4-Trifluoromethylphenyl)-N-[4-
(methoxycarbonyl)benzyl]-N-(l-methylpiper idin-4-yl) acetamide (50ELH20A).
[0347J Starting materials: 50ELH19 (0.20 g, 0.76 mmol, 1.0 eq.), 50ELH12'(0.169 g, l.O eq.).
[03481 Product: Yield 108.9 mg (32%), UV/MS 100/100 (M+ 448), r, (A, MS) 3.327, Rf 0.3 (5% MeOH/CH2Cl2). 1H-NMR (400 MHz3DMSO, rotamers 56/44) δ 10.7 and 10.4 (2brs, IH), 7.96-7.28 (m, 8H), 4.70 and 4.51 (2s, 2H)3 4.30 (brt, IH)5 4.06 and 3.69 (2s, 2H)3 3.83 and 3.81 (2s, 3H), 3.00 (m, 2H), 2.63 (m, 3H), 2.05 (brt, J=12 Hz, 2H)5 1.69 (brt, J=12 Hz5 2H). 13C-NMR (CDCl3) δ 171.9; 166.7; 142.9; 138.5; 130.7; 130.1 ; 129.7; 126.2; 125.9; 55.2; 52.5; 49.2; 47.4; 41.2; 32.1; 26.6; 22.9; 14.3.
Example 64 - 2-Phenyl-N-[4-rmethoxycarbonyl')benzyl1-N-('l-methylpiperidin-4-yl) acetamide (50ELH20B)
[03491 Procedure as 50ELH14B
[0350] Reaction-Step 2: 2-Phenyl-N-[4-(methoxycarbonyl)benzyl]-N-(l- methylpiperidin-4-yl) acetamide (50ELH20B)
[0351] Starting materials: 50ELH19 (0.2 g, 0.76 mmol, 1.0 eq.), phenylacetylchloride (0.117 g, 1.0 eq.).
[0352] Product: Yield 82.5 g (29%), UV/MS 100/100 (M+ 381), rt (A, MS) 2.652, Rf 0.25 (30% MeOH/CH2Cl2). 1H-NMR (400 MHz, CDCl3) δ 12.2 (brs, IH), 8.00 (d, J=7.4, 2H), 7.4-7.2 (m, 4H), 7.08 (d, J=7.4, 2H), 4.89 (brt, IH), 4.62 (s, 2H)5 3.90 (s, 3H), 3.56 (s, 2H), 3.42 (d, J=I LO5 2H), 2.84 (q, J=I LO, 2H), 2.68 (d, J=3.65 3H), 240 (q, J=I LO5 2H), 1.77 (brd, J=I LO, 2H). 13C-NMR δ 173.0; 168.0; 143.3; 136.7; 130.6; 129.0; 127.4; 125.9; 54.5; 52.4; 48.8; 43.6; 41.4; 26.3.
Example 65 - 2-(4-Chlorophenvπ-N-r4-rmethoxycarbonvπbenzyl]-N-(l-methylpiperidin- 4-vn acetamide C50ELH20O.
[0353] Procedure as 50ELH14B.
[0354] Reaction-Step 2: 2-(4-Chlorophenyl)-N-[4-(methoxycarbonal)benzyl]- N-(l-methylpiperidin-4-yl ) acetamide (50ELH20C).
[0355] Starting materials: 50ELH19 (0.2 g, 0.76 mmol, 1.0 eq.), A- chlorophenylacetylchloride (0.131 g, 1.0 eq.).
[0356] Product: Yield 79.2 g (26%), UV/MS 100/96 (M+ 399), rt (A, MS) 2.333. 1H-NMR (400 MHz, DMSO5 rotamers 62/38) δ 10.8 and 10.60 (2brs, IH)5 7.95 and 7.85 (2d, J=8.6, 2H), 7.4 and 7.28 (2d5 2H)5 7.35 and 7.14 (2m5 4H)5 4.67 and 4.50 (2s, 2H), 4.29 (m, IH), 3.93 and 3.84 (2s, 2H)5 3.81 (s, 3H)5 3.21 (d, J=I 1.95 2H)5 3.00 (d, J=I 1.9, 2H)5 2.63 (s, 3H), 2.06 (m, 2H)5 1.68 and 1.56 (d5 J=I 1.95 2H). 13C-NMR (CDCl3) δ 172.6; 166.7; 163.4; 161.0; 143.0; 130.7; 130.6; 130.5; 126.0; 115.9; 115.7; 54.7; 52.4; 48.9; 46.9; 44.0; 40.4; 26.4.
Example 66 - 2-(4-MethoxyphenylVN-[4-('methoxycarbonyl*)benzyll-N-('l- methylpiperidin-4-yl ) acetamide f50ELH20D).
[0357] Procedure as 50ELH14B.
[0358] Reaction-Step 2: 2-(4-Methoxyphenyl)-N-[4-
(methoxycarbonyl)benzyl]-N-(l-methylpiperidin-4-y 1) acetamide (50ELH20D).
[0359] Starting materials: 50ELH19 (0.2 g, 0.76 mmol, 1.0 eq.), A- methoxyphenylacetylchloride (0.140 g, 1.0 eq.).
[0360] Product: Yield 108.6 g (26%), UV/MS 100/99 (M+ 410), r, (A, MS) 2.280. 1H-NMR (400 MHz, CDCl3) δ 12.38 (brs, IH), 8.00 (d, J=7.2, 2H), 7.28 (d, J=7.2, 2H), 7.00 (d, J=7.2, 2H), 6.79 (d, J=7.2, 2H), 4.88 (bit, IH), 4.61 (s, 2H), 3.90 (s, 3H), 3.75 (s, 3H), 3.42 (brd, J=I 0.7, 2H), 2.84 (q, J=I 0.7, 2H)5 2.68 (d5 J=3.6, 3H), 2.40 (brq, J=10.7, 2Hl)5 1.75 (d, J=10.75 2H). 13C-NMR δ 173.0; 166.8; 159.0; 143.3; 130.5; 129.9; 129.8; 126.3; 125.9; 114.5; 55.5; 54.7; 52.4; 48.7; 46.7; 43.6; 40.6; 32.1; 26.3; 22.9; 14.3. Example 67 - 2-f4-TMemylphenylVN-r4-fmefooxycarbonvnbenzyl1-N-O- methylpiperidin-4-yl } acetamide (50ELH23")
[0361] Procedure as 50ELH14B.
10362] Reaction-Step 2: l-Phenyl-N-[2-(4-methylphenyl)ethyl]-N-(l- methylpiperidin-4-yl) amide (50ELH23).
[0363] Starting materials: 4-(2-Phenylethyl)amino-l-methyIpiperidine (0.20 g, 0.86 mmol, 1.0 eq.), benzoylchloride (0.158 g, 1.0 eq.).
[0364] Product: Yield 159 mg (50%), UV/MS 100/100 (M+ 337), rt (A, MS) 3.289, Rf 0.55 (10% MeOH/CH2Cl2). 1H-NMR (400 MHz, DMSO (80° C)) δ 10.9 (brs, IH), 7.44 (s, 2H)5 7.34 (d, J=3.0 Hz, 2H), 7.04 (d, J=7.0 Hz, 2H), 6.95 (brs, 2H), 4.00 (brs, IH), 3.40 (d, J=4.2 Hz3 2H), 3.35 (d, J=4.2 Hz5 2H), 2.95 (brs5 2H), 2.77 (t, J=3.2 Hz, 2H), 2.40 (q, J=6.4 Hz, 2H)5 2.24 (s, 3H) 1.83 (d, J=6.4 Hz5 2H). 13C-NMR (CDCl3) δ 171.6; 138.1; 136.3; 136.0; 129.8; 129.6; 129.1; 129.1; 126.7; 53.6; 52.4; 46.1; 42.9; 35.9; 27.3; 21.1.
Example 68 - 2-r4-Methoxyphenyl')-N-('3-phenyl-l-propyl')-N-('l-methylpiperidin-4-yl) acetamide (50ELU65)
[0365] Procedure as 50ELH 14B.
[0366] Reaction-Step 1 : 4-(3-Phenylaminopropyl)piperidine (50ELH59)
[0367] Starting materials: l-Methyl-4-piperidone (1.1 ml, 7.4 mmol, 1.0 eq.), 3-phenylpropylamine (1.35 g, 1.0 eq.).
[0368] Product. UV/MS 100/94 (M+ 233), rt (A5 MS) 3.534.). 1H-NMR (400 MHz5 CDCl3) δ 7.28-7.12 (m, 5H)5 3.40 (brs, IH), 2.84 (dt, J=12.3 and 3.5 Hz5 2H)5 2.94 (g, J=7.0 Hz, 4H), 2.51 (m, IH), 2.27 (s, 3H), 2.05 (brt, J= 12.3 Hz, 2H)5 1.82 (m5 2H)5 1.44 (m5 2H).
[0369] Reaction-Step 2: 2-(4-Methoxyphenyl)-N-(3-phenyl-l-propyl)-N-(l- methylpiperidin-4-yl) acetamide (50ELH65)
[0370] Starting materials: 50ELH59 (0.50 g, 2.2 mmol, 1.0 eq.), 4- methoxyphenylacetylchloride (0.398 g, 1.0 eq.).
[0371] Product: Yield 153 mg (43%), UV/MS 100/100 (M+ 381), rt (A5 MS) 2.938. 1H-NMR (400 MHz, DMSO, rotamers 55/45) δ 11.0 and 10.90 (2brs, IH)5 7.30- 7.10 (m, J=7.9 Hz5 6H), 6.97 (d, J=7.9 Hz, IH)5 4.22 and 4.06 (2dt, dH), 3.70 (s, 3H), 3.35 (t, J=10.4 Hz, 2H), 3.15 (m, 2H), 3.00 (q, J=10.4 Hz, 2H)5 2.66 (d, 3H), 2.52 (q, J=7.9 Hz, 2H), 2.17 (brq, J=12 Hz, 2H) 1.73 (m, 2H), 1.70 and 1.52 (2d, J=12 Hz, 2H). 13C-NMR (DMSO) δ 171.3; 171.0; 158.6; 142.2; 141.7; 130.0; 129.0; 128.0; 128.5; 128.2; 126.6; 114.5; 55.7; 55.7; 53.5; 53.3; 50.1; 44.5; 42.9; 41.9; 33.7; 33.1; 32.9; 31.4; 27.8; 26.8.
Example 69 - 2-(4-Methoxyphenyl)-N-r2-('4-methylphenyπethyl)-N-('l-methylpiperidin-4- vD acetamide (50ELH68)
[0372] Procedure as 50ELH14B
[0373] Reaction-Step 1 : 4-[2-(4-Methylphenyl)ethylamino]-piperidin (50ELH58)
[0374] Starting materials: l-Methyl-4-piperidone (1.1 ml, 7.4 mmol, 1.0 eq.), 2-(4-methylphenyl)ethylamine (1.0 g, 1.0 eq.).
[0375] Product: UV/MS 100/91 (M+ 233), rt (A, MS) 3.933.). 1H-NMR (400 MHz, CDCl3) δ 7.4 (s, 5H), 3.27 (brs, IH), 2.84 (d, J=7.0 Hz5 4H)5 2.75 (m5 2H)5 2.54 (m5 IH), 2.29 (2xs, 6H), 2.10 (brt, J=12.3 Hz, 2H), 1.86 (brd, 2H)5 1.45 (m, 2H).
[0376] Reaction-Step 2: 2-(4-Methoxyphenyl)-N-[2-(4-methylphenyl)ethyl)- N-(l-methylpiperidin-4-yl) acetamide (50ELH68)
[0377] Starting materials: 50ELH58 (0.30 g, 1.3 mmol, 1.0 eq.)5 4- methoxyphenylacetylchloride (0.238 g, 1.0 eq.).
Product: Yield 125 mg (26%), UV/MS 100/99 (M+ 381), rt (A, MS) 3.156. 1H-NMR (400 MHz3 DMSO, rotamers 50/50) δ 11.0 and 10.90 (2brs, IH), 7.25-7.04 (m, J=8.7 Hz, 6H)5 6.87 and 6.84 (2d, J=8.7 Hz5 2H), 4.30 and 4.09 (2dt, J=I 1.5 Hz5 dH)5 3.73 and 3.58 (2s, 2H), 3.71 and 3.70 (2s, 3H), 3.35 (m, (Underneath waterpeak) 3H), 3.24 (m, IH), 3.02 (m, J=I 1.5 Hz, 2H), 2.80-2.62 (m, 5H), 2.32 and 2.20 (2q, J=I 1.5Hz, 2H), 2.26 and 2.24 (2s, 3H) 1.78 and 1.49 (2d, J=I 1.5 Hz, 2H). 13C-NMR (DMSO) δ 171.5; 171.2; 158.6; 136.8; 136.2; 136.0; 135.8; 130.7; 130.5; 129.7; 129.6; 129.4; 129.2; 128.4; 128.3; 114.5; 55.8; 55.7; 53:3; * 53.3; 52.2; 50.2; 46.8; 43.9; 42.9; 36.8; 35.2; 27.6; 26.8; 21.3.
Example 70 - 2-r4-MethoxyDhenylVN-r2-f2-thienyl')ethvη-N-π-methylpiperidin-4-vn acetamide (50ELH71 A)
[0378] Procedure as 50ELH14B [0379] Reaction-Step 1 : 4-[2-(2-Thienyl)ethylamino]ρiperidin (50ELH67A)
[0380] Starting materials: l-Methyl-4-piperidone (0.5 g, 4.4 mmol, 1.0 eq.), thiophene-2-ethylamine (0.563 g, 1.0 eq.).
[0381] Product: UV/MS 94/93 (M+ 225).
[0382] Reaction-Step 2: 2-(4-Methoxyphenyl)-N-[2-(2-thienylethyl]-N-(l- methylpipieridin-4-yl) acetamide (50ELH71A)
[0383] Starting materials: 50ELH67A (0.243 g, 1.08 mmol, 1.0 eq.) 4- methoxyphenylacetylchloride (0.2 g, 1.0 eq.).
[0384] Product: Yield 80.7 mg (33%), UV/MS 100/100 (M+ 373), rt (A, MS) 2.613. 1H-NMR (400 MHz, DMSO, rotamers 50/50) δ 10.8 and 10.6 (2brs, IH)5 7.36 and 7.31 (2d, J=4.7 Hz. IH), 7.20 and 7.06 (2d, J=8.3 Hz, 2H)3 7.00-6.92 (m, J=4.7 and 2.8 Hz, 2H), 6.87 and 6.40 (2d, J=8.3 Hz3 2H)3 4.22 and 4.08 (2dt, J=12.2 Hz, IH), 3.71 (s, 3H)3 3.70 (S3 2H)3 3.46-3.30 (m3 4H), 3.10-2.90 (m, 4H), 2.67 (m, 2H), 2.28 and 2.12 (2q, J=12 Hz3 2H)3 1.80 and 1.50 (2d, J=12 Hz, 2H). 13C-NMR (DMSO) δ 172.5; 158.9; 139.6; 130.0; 129.6; 126.8; 124.5; 114.5; 55.5; 54.7; 49.3; 45.8; 43.8; 41.3; 31.9; 29.9
Example 71 - 2-(4-Methoxyphenyl')-N-f2-('4-nitrophenyl')ethvn-N-fl-methylpiperidin-4- yl) acetamide (50ELH71O
[0385] Procedure as 50ELH14B
Reaction-Step 1 : 4-[2-(4-nitrophenyl)ethylamino]-piperidin (50ELH67C)
[0386] Starting materials: l-Methyl-4-piperidone (0.5 g, 4.4 mmol, 1.0 eq.), 4- nitrophenyl-2-ethylamine (0.897 g, 1.0 eq.).
[0387] Product: UV/MS 96/89 (M+ 264), r, (A, MS) 3.264.
[0388] Reaction-Step 2: 2-(4-Methoxyphenyl)-N-[2-(4-nitrophenyl)ethyl]-N- (l-methylpiperidin-4-yl) acetamide (50ELH71 A)
[0389] Starting materials: 50ELH67C (0.285 g3 1.08 mmol, 1.0 eq.)3 4- methoxyphenylacetylchloride (0.2 g, 1.0 eq.).
[0390] Product: Yield 130.9 mg (30%), UV/MS 100/100 (M+ 412), rt(A, MS) 2.219. 1H-NMR (400 MHz, DMSO, rotamers 50/50) δ 10.8 and 10.6 (2brs, IH), 8.17 and 8.12 (2d, J=8.6 Hz, 2H)3 7.58 and 7.48 (2d3 J=8.6 Hz3 2H), 7.2 and 7.1 (2d3 J=8.6 Hz3 2H), 6.87 and 6.40 (2d3 J=8.6 Hz3 2H), 4.25 and 4.10 (2dt, J=12 Hz, IH)3 3.72 (s, 3H), 3.70 (s, 2H), 3.48-3.30 (m, 4H), 3.10-2.84 (m, 4H), 2.69 and 2.67 (2d, J=4.7 Hz, 3H), 2.34 and 2.15 (2q, J=13.2 Hz5 2H), 1.79 and 1.47 (2d, J=13.2 Hz, 2H).
Example 72 - 2-(4-Methoxyphenyl)-N-('2-thienylmethylVN-d-methylpiperidin-4-vπ acetamide (50ELH73A')
[0391] Procedure as 50ELH14B.
[0392] Reaction-Step 1: 4-[(2-Thienylmethyl)amino]-l-methylpiperidine (50ELH66A)
[0393] Starting materials: l-Methyl-4-piperidone (0.5 g, 4.4 mmol, 1.0 eq.), 2- thienylmethylamine(0.52 g, 1.0 eq.).
[0394] Product: UV/MS 77/86 (M+ 211 ), rt (A3MS) 2.739.
[0395] Reaction-Step 2: 2-(4-Methoxyphenyl)-N-(2-thienylmethyl)-N-(l- methylpiperidin-4-yl) acetamide (50ELH73A)
[0396] Starting materials: 50ELH66A (0.228 g, 1.08 mmol, 1.0 eq.), 4- methoxyphenylacetylchloride (0.2 g, 1.0 eq.).
[0397] Product: Yield 178.4 mg (50%), UV/MS 100/98 (M+ 359), rt (A, MS) 3.117. 1H-NMR (400 MHz, DMSO) δ 10.9 and 10.6 (2brs, IH), 7.47 and 7.32 (2d, J=4.5 Hz, IH), 7.20 and 7.03 (2d, J=8.4 Hz, 2H)5 7.03 and 6.98 (2m, IH), 6.87 (m, 3H)5 4.70 and 4.57 (2s, 2H), 4.42 and 4.16 (2t, J=I 1.9 Hz5 IH)5 3.77 and 3.60 (2s, 2H), 3.51 (s, 3H), 3.15 (m, 2H), 2.98 (m, J=I 1.9 Hz, 2H), 2.65 (2d, J=4.5 Hz5 3H), 2.25 and 2.17 (2q, J=I 1.9 Hz5 2H)5 1.69 and 1.44 (2d5 J=I 1.9 Hz, 2H). 13C-NMR (DMSO) δ 171.4; 158.6; 143.2; 130.7; 128.1 ; 126.6; 126.3; 125.9; 114.5; 55.7; 53.3; 52.6; 50.0; 42.8; 27.7; 26.8.
Example 73 - 2-(4-MethoxyphenylVN-ffυrrarylVN-π-methylpiperidin-4-vO acetamide (50ELH73B).
[0398] Procedure as 50ELH14B.
[0399] Reaction-Step 1: 4-(Furfurylamino)-l-methylpiρeridin (50ELH66B) Starting materials: l-Methyl-4-piperidone (0.5 g, 4.4 mmol, 1.0 eq.)5 Furfurylamine (0.43 g, 1.0 eq.).
[0400] Product: UV/MS 77/92 (M+ 195), rt (A, MS) 2.812.). Reaction-Step 2: 2-(4-Methoxyphenyl)-N-(fiirfuryl)-N-(l-methylpiperidin-4-yl) acetamide (50ELH73B). [0401] Starting materials: 50ELH66B (0.21 g, 1.08 mmol, 1.0 eq.), 4- methoxyphenylacetylchloride (0.2 g, 1.0 eq.).
[0402] Product: Yield 134 mg (36%), UV/MS 100/99 (M+ 343), r, (A5 MS) 2.401. 1H-NMR (400 MHz, DMSO5 rotamers 57/43) δ 10.95 and 10.75 (2brs, IH)5 7.63 and 7.48 (s5 IH)5 7.18 and 7.06 (2d, J=7.7 Hz, 2H), 6.85 (t, J=7.7 Hz, 2H)5 6.44 and 6.33 (2d5 J=7.7 Hz5 IH), 6.37 and 6.11 (2s, IH) 4.5 and 4.34 (2s, 2H)5 4.42 and 4.18 (2dt, J=I 1 and 2 Hz, IH)5 3.75 and 3.65 (2s, 2H) 3.70 (s, 3H), 3.33 (hidden, 2H)5 3.0 (q, 2H), 2.64 (d, J=4.7 Hz, 3H)5 2.15 (dq5 J=I 1 and 2 Hz, 2H)5 1.65 and 1.50 (2d, J=I 1 Hz, 2H).
Example 74 - 2-(2-thienyl)-N-(4-methylphenylmethyl)-N-n-methylpiperidin-4-yl) acetamide (50ELH82)
[0403] Procedure as 50ELH14B
[0404] Reaction-Step 2: 2-(2-thienyl)-N-(4-methylphenylmethyl)-N-(l- methyIpiperidin-4-yl) acetamide (50ELH82)
[0405] Starting materials: 50ELH25 (0.30 g5 1.38 mmol, 1.0 eq.), thiophene-2- acetylchlorid (0.22 g, 1.0 eq.).
[0406] Product: Yield 235 mg (62%), UV/MS 97/93 (M+ 343), rt (A5 MS) 2.795. 1H-NMR (400 MHz5 DMSO5 rotamers 54/46) δ 10.8 and 10.60 (2brs, IH), 7.4 and 7.35 (2d5 IH)3 7.2-6.76 (m5 6H), 4.55 and 4.4 (2s5 2H)5 4.49 and 4.26 (2dt, J=I 1 and 2 Hz, 2H), 4.15 and 3.79 (2s, 2H), 3.32 (d, J=I l Hz, 2H), 2.99 (q, 2H), 2.63 (s, 3H), 2.27 and 2.23 (2s, 3H), 2.09.(q, J=I 1 Hz, 2H), 1.66 and 1.55 (2d, J=I 1 Hz, 2H).
Example 75 - 2-(4-Meτhoxyphenvπ-N-(4-methylbenzyl)-N-(l-cyclopentylpiperidin-4-vπ acetamide (42ELH75)
[0407] Procedure as for 42ELH80, except that the reaction was run at 60° C. for 3 days.
[0408] Starting materials: 50ELH87 (0.25 g, 0.71 mmol, 1.0 eq.), Cyclopentylbromide (0.288 g, 3.0 eq.).
[0409] Product: Yield 91.2 mg (34%), UV/MS 88/93 (M+ 421), rt (A, MS) 4.450. Example 76 - 2-r4-MethoxyphenylVN-f4-methylbenzylVN-α-r3-d.3-dihvdro-2H- benzimidazol -2-one-l-v0propyI')piρeridine-4-yl) acetamide (50ELH89).
[0410] 50ELH87 (0.05 g, 0.14 mmol, 1 eq.) was transferred to a 4 ml vial and dissolved in 1 ml of acetonitrile. Then, l-(3-chloropropyl)-l,3-dihydro-2H-benzimidazol- 2-one (0.032 g, 1.1 eq.)5 sodium carbonate (0.022 g, 1.1 eq.) and KI (one crystal) were added and the vial was sealed and shaken for 20 h at 82° C. The mixture was extracted with distilled water (pH~10, sodium carbonate) and dichloromethane (3 times) the organic layers were dried with sodium sulfate and concentrated. The title compound was purified by HPLC and evaporated to dryness, forming a trifluoroacetic acid salt. Yield 8.8 mg (12%). UV/MS 100/100 (M+ 527), rt(A, MS) 2.851.
Example 77 - 2-f4-Methoxyphenyl')-N-r4-methylbenzvD-N-ri-('2-methylthiazol-4- ylmethyD piperidin-4-yl") acetamide (63ELH1 A).
[0411] 50ELH87 (0.3 g, 0.852 mmol, 1.0 eq) and 4-(chloromethyl)-2- methylthiazole hydrochloride (0.235 g, 1.5 eq) were added to a 7 ml vial and dissolved in acetonitrile (3 ml). Potassium carbonate (141.3 g, 1.2 eq) and a crystal of potassium iodide were added and the vial was sealed and shaken for 20 h at 82° C. The reaction mixture was extracted with distilled water (made basic by potassium carbonate, pH 10) and dichloromethane. The crude product was dried with sodium sulfate and concentrated. After purification by HPLC the product was converted into the hydrochloride salt by dissolving the free base in 1 ml dichloromethane and adding 1 eq. HCl in ether (2M). This mixture was added drop-wise to an excess of heptane where the product precipitated. The solvent was removed by evaporation leaving a white powder as the product, yield 83.8 mg (21%), UV/MS 100/90 (M+ 463), rt(B, MS) 11.82.
Example 78 - 2-f4-Memoxyphenyl>N-(2-4-(fluorophenvD ethylVN-π-methylpiperidin-4- yl) acetamide r50ELH93A)
[0412] Procedure as 50ELH14B.
[0413] Reaction-Step 1: 4-[2-4-(Fluorophenyl)ethylamino]-l-methylpiperidine (50ELH92A)
[0414] Starting materials: l-Methyl-4-piperidone (0.3 g, 2.65 mmol, 1.0 eq.), 4-(fluorophenyl)ethylamine (0.369 g, 1.0 eq.).
[0415] Product: UV/MS 60/92 (M+ 237), rt (A5MS) 3.422. [0416] Reaction-Step 2: 2-(4-Methoxyphenyl)-N-(2-4-(fluorophenyt)ethyl)-N- (l-methylpiperidin-4-yl) acetamide (50ELH93A)
[0417] Starting materials: 50ELH92A (0.625 g, 2.65 mmol, 1.0 eq.), 4- methoxyphenylacetylchloride (0.488 g, app. 1.0 eq.).
[0418] Product: Yield 181 mg (18%), UV/MS 87/97 (M+ 385), r, (A, MS) 2.783. Rf 0.8 (10% MeOH/CH2Cl2). 1H-NMR (400 MHz5 DMSO, rotamers 50/50) δ 10.9 (brs, IH)5 7.56-6.8 (m, 8H), 4.26 and 4.02 (2brt, 2H)5 3.70 and 3.95 (2s, 3H), 3.59 and 3.57 (2s, 2H), 3.4-3.15 (m, 5H)5 2.96-2.66 (m, 5H)5 2.62 and 2.56 (2s5 3H)5 2.29 and 2.10 (2q, 2H), 1.73 and 1.41 (2d. 2H). 13C-NMR (DMSO) δ 172.5; 171.4; 171.3; 162.9; 162.7; 160.5; 160.3; 158.9; 158.6; 136.1; 136.1; 135.3; 131.4; 131.3; 131.1; 131.0; 131.0; 130.6; 130.5; 128.4; 128.4; 126.9; 115.9; 115.8; 115.7; 115.6; 114.5; 55.7; 53.7; 53.5; 52.7; 52.3; 50.7; 46.7; 43.8; 43.2; 43.0; 36.3; 34.7; 27.9; 26.9.
Example 79 - 2-(4-MethoxyDhenvD-N-r2-f2.5-dimethoxyphenyl)ethyll-N-π- methylpiperidin-4 -yl) acetamide (50ELH93C)
[0419] Procedure as 50ELH14B. A small amount was purified by HPLC and evaporated to dryness, forming the trifluoroacetic acid salt.
[0420] Reaction-Step 1: 4-[2-(2,5-dimethoxyphenyl)ethylamino]-l- methylpiperidine (50ELH92A)
[0421] Starting materials: Methyl-4-piperidone (0.3 g, 2.65 mmol, 1.0 eq.), 2,5-(dimethoxyphenyl)ethylamine (0.481 g, 1.0 eq.).
[0422] Product: UV/MS 81/90 (M+ 279), rt(A, MS) 2.868.
[0423] Reaction-Step 2: 2-(4-Methoxyρhenyl)-N-[2-(2. 5- dimethoxyphenyl)ethyl]-N-(l-methylpiperidin-4-yl) acetamide (50ELH93C)
[0424] Starting materials: 50ELH93C (0.737 g, 2.65 mmol, 1.0 eq.), 4- methoxyphenylacetylchloride (0.488 g, app. 1.0 eq.).
[0425] Product: UV/MS 82/100 (M+ 427), r, (B, MS) 8.44. Rf 0.8 (10% MeOH/CH2Cl2).
Example 80 - 2-r4-Methoxyphenyl)-N-r2-f2.4-dichlorophenvnethvn-N-(l- methylpiperidin-4- yl) acetamide r50ELH93D') [0426J Procedure as 50ELH14B, but purified by HPLC and evaporated to dryness forming the trifluoroacetic acid salt.
[0427] Reaction-Step 1: 4-[2-(2,4-Dichlorophenyl)ethylamino]-l- methylpiperidine (50ELH92D)
[0428] Starting materials: l-Methyl-4-piperidone (0.3 g, 2.65 mmol, 1.0 eq.)5 2,5-(dichlorophenyl) ethylamiπe (0.50 g, 1.0 eq.).
[0429] Product: UV/MS 82/92 (M+ 287), rt (A, MS) 4.875.
[0430] Reaction-Step 2: 2-(4-Methoxyphenyl)-N-[2-(2,4- dichlorophenyl)ethyl]-N-(l-methylpiperidin-4 -yl) acetamide (50ELH93D)
[0431] Starting materials: 50ELH93D (0.76 g, 2.65 mmol, 1.0 eq.), 4- methoxyphenylacetylchloride (0.488 g, app. 1.0 eq.).
IO432] Product: UV/MS 100/96 (M+ 435), rt (A, MS) 4.415. Rf 0.8 (10% MeOH/CH2Cl2).
Example 81 - 2-(4-Methoxyphenyl')-N-r2-(3-chlorophenyl')ethvn-N-π -methylpiperidin-4- vD acetamide fSOELHgSE')
[0433] Procedure as 50ELH14B, but purified on HPLC and evaporated to dryness forming the trifluoroacetic acid salt.
[0434] Reaction-Step 1: 4-[(3-Chlorophenyl)ethyl)amino]-l-methylpiperidine (50ELH92E)
[0435] Starting materials: l-Methyl-4-piperidone (0.3 g, 2.65 mmol, 1.0 eq.), 3-(chlorophenyl) ethylamine (0.413 g, 1.0 eq.).
[0436] Product: UV/MS 86/88 (M+ 253), rt (A,MS) 3.175.
[0437] Reaction-Step 2: 2-(4-Methoxyphenyl)-N-[2-(3-chlorophenyl)ethyl]-N- (l-methylpiperidin-4-yl) acetamide (50ELH93E)
[0438] Starting materials: 50ELH93E (0.67 g, 2.65 mmol, 1.0 eq.)5 4- methoxyphenylacetylchloride (0.488 g, app. 1.0 eq.).
[0439] Product: UV/MS 100/100 (M+ 401), rt (A, MS) 3.464. Rf 0.8 (10% MeOH/CH2Cl2). Example 82 - 2-r4-MethoxyDhenvn-N-|'2-f4-methoxyphenyl')ethyll-N-ri- methylpiperidin-4-yl) acetamide (50ELH95B)
[0440] Procedure as 50ELH14B. Purified by HPLC and evaporated to dryness forming the trifluoroacetic acid salt.
[0441] Reaction-Step 1: 4-[(4-MethoxyphenyI)ethyl)amino]-l- methylpiperidine (50ELH94B)
[0442] Starting materials: l-Methyl-4-piperidone (0.3 g, 2.65 mmol, 1.0 eq.), 4-methoxyphenylethylamine (0.40 g, 1.0 eq.).
[0443] Product: UV/MS 74/87 (M+ 249), rt (A5MS) 2.935.
[0444] Reaction-Step 2: 2-(4-Methoxyphenyl)-N-[2-(4-methoxyphenyl)ethyl]- N-(l-methylpiperidin-4-yl ) acetamide (50ELH95B)
[0445] Starting materials: 50ELH94B (0.657 g, 2.65 mmol, 1.0 eq.), 4- methoxyphenylacetylchloride (0.488 g, app. 1.0 eq.).
[0446] Product: UV/MS 100/100 (M+ 397), r, (A, MS) 2.389. Rf 0.8 (10% MeOH/CH2Cl2).
Example 83 - 2-r4-Methoxyphenyl)-N-f2-r3-fluorophenvπethyl]-N-('l-methylpiperidin-4- yl> acetamide (SOELmδO')
[0447] Procedure as 50ELH14B. Purified on HPLC and evaporated to dryness, forming the trifluoroacetic acid salt.
[0448] Reaction-Step 1 : 4-[2-((3-Fluorophenyl)ethyl)amino]-l- methylpiperidine (50ELH94D)
[0449] Starting materials: l-Methyl-4-piperidone (0.3 g, 2.65 mmol, 1.0 eq.), 3-fluorophenylethylamine (0.369 g, 1.0 eq.).
[0450] Product: UV/MS 74/89 (M+ 237), rt (A5MS) 2.946.
[0451] Reaction-Step 2: 2-(4-methoxyphenyl)-N-[2-(3-fluorophenyl)ethy I]-N- (l-methylpiperidin-4-yl) acetamide (50ELH95D)
[0452] Starting materials: 50ELH94D (0.625 g, 2.65 mmol, 1.0 eq.), 4- methoxyphenylacetylchloride (0.488 g, app. 1.0 eq.).
[0453] Product: UV/MS 100/95 (M+ 385), rt (A, MS) 2.946. Rf 0.8 (10% MeOH/CH2Cl2). Example 84 - 2-(4-ethoxyphenyl')-N-f2-('4-fluorophenyπethyl]-N-π-methylpiperidin-4- yli acetamide (63ELH20)
[0454] Reaction Step, 1 : 4-Ethoxyphenylacetic acid chloride(63ELH 19)
[0455] 4-Ethoxyphenylacetic acid (0.5 g, 2.8 mmol) was transferred to a 7 ml vial and dissolved in thionylchloride (3 ml). The reaction mixture was shaken at 70° C. for 21/2 hours. Thionylchloride was evaporated off and the resulting product was used unpurified.
[0456] Reaction Step 2: 2-(4-Ethoxyphenyl)-N-[2-(4-fluorophenyl)ethyl]-N- (l-methylpiperidin-4-yl) acetamide (63ELH20)
[0457] 63ELH17 (0.11 g, 0.47 mmol) was transferred to a 4 ml vial and dissolved in dichloromethane. 63ELH19 (0.084 mg, 1 eq.) was added and the vial was sealed and the reaction shaken for 20 h. The product was extracted in distilled water (made basic with potassium carbonate, pH 10) and dichloromethane. Dried with sodium sulfate and concentrated. Purified by HPLC. The extraction, drying and concentration was repeated and the product re-dissolved in dichloromethane (1 ml) and HCl (1 eq., 2 M in ether) was added. The mixture was added drop-wise to an excess of heptane whereupon the salt precipitated. Yield 33.4 mg (18%), UV/MS: 92/100 (M+ 399), tr (B, MS) 10.38.
Example 85 - 2-f4-EthoxyphenylVN-(4-fluorobenzyl)-N-π-methylpiperidin-4-yl) acetamide (63ELH21)
[0458] 50ELH4 (0.11 g, 0.49 mmol, 1.0 eq.) was transferred to a 4 ml vial and dissolved in dichloromethane. 63ELH19 (0.089 mg, 1.0 eq.) was added and the vial was sealed and the reaction shaken for 20 h. The product was extracted in distilled water (made basic with potassium carbonate, pH 10) and dichloromethane. Dried with sodium sulfate and concentrated. Purified by HPLC. The extraction, drying and concentration was repeated and the product dissolved in dichloromethane (1 ml) and HCl (1 eq., 2 M in ether)is added. This mixture was added drop-wise to an excess of heptane whereupon the salt precipitated. Yield 31.1 mg (16%), UV/MS: 94/100 (M+ 385), tr (A, MS) 2.573.
Example 86 - N-f^-methylphenvDmethylVN-f l-methylpiperidin-4-ylV2-(3-hydroxy-4- methox vphenvDacetamide C57MBT12B~)
[0459] N-((4-methylphenyl)methyl)-4-amino- 1 -methylpiperidine (50ELH25) (105 mg, 0.48 mmol) and 3-hydroxy-4-methoxyphenylacetic acid (88 mg, 0.48 mmol) were dissolved in DMF (10 ml). Diisopropylethylamine (DIEA, 250μL5 1.44 mmol) was added followed by bromo-tris-pyrrolidino-phosphonium hexafluorophosphate (PyBrOP, 336 mg, 0.72 mmol), and the mixture was stirred at r.t for 1 h. Water (50 mL) was added, and the reaction mixture was extracted with EtOAc (2 X 50 mL). Drying by Na2SO4 and concentration yielded 514 rng crude material, which was purified by flash chromatography (0-30% MeOH in CH2Cl2). This gave 105 mg (57%)of the title compound as a white solid. Rf =0.20 (10% MeOH in CH2Cl2). HPLC-MS (method A) showed MH+ =383. U V/MS(%)= 100/92. 1H-NMR (400 MHz, CD3OD, Rotamers 52:48): δ 7.18-6.58 (m, 7H)5 4.53 (s, 2H), 4.31 and 3.97 (2m, IH), 3.82 and 3.81 (2s, 3H), 3.80 and 3.55 (2s, 2H), 3.04 and 2.85 (2m, 2H), 2.41 and 2.32 (2s, 3H), 2.35 and 2.12 (2m, 2H), 2.29 and 2.27 (2s, 3H), 1.83 and 1.74 (2m, 2H), 1.72 and 1.33 (2m, 2H)
Example 87 - N-(T4-methylDhenyl')methvπ-N-fl-methylpiperidin-4-ylV2-f3.4- dihvdroxyphenv Hacetamide f57MBT24B')
[0460] N-((4-methylphenyl)methyl)-N-(l-methylpiperidine-4-yl)-2-(3- hydroxy-4-metho xyphenyl)acetamide (57MBT12B) (52 mg, 0.136 mmol) was dissolved in CH2Cl2 (1 mL) and cooled to -78 ° C. Boron tribromide (IM in CH2Cl2, 204μl, 0.204 mmol) was added dropwise and the cooling bath was removed. After stirring for 2 h, methanol (2 mL) was added and the mixture was evaporated. The resulting oil was purified by preparative HPLC to give 24 mg (48%) of the title compound as a white solid. HPLC-MS (method A) showed MH+ =369. UV/MS(%)=100/97. IH-NMR (400 MHz, CD3OD, Rotamers 33:67): δ 7.19-6.47 (m, 7H), 4.54 and 4.53 (2s, 2H), 4.23 (m, IH), 3.83 and 3.58 (2s, 2H), 3.46 and 3.40 (2br d, J=12 Hz, 2H), 3.02 and 2.95 (2br t, J=12 Hz, 2H)5 2.79 (s, 3H), 2.33 and 2.28 (2s, 3H), 2.17 and 1.84 (2dq, J=4, 12 Hz, 2H), 1.87 and 1.48 (2br d, J=I 2 Hz5 2H)
Example 88 - N-(r3-hvdroxy-4-methylphenyl)methvπ-N-d»methylpiperidin-4-yl)-2-(4- methox vphenvQacetamide T57MBT54B)
[0461] N-((4-methoxyphenyl)methyl)-4-amino-l-methylpiperidine (1 g, 4.27 mmol) was dissolved in 4% formic acid in methanol (60 mL). 10% Pd/C (1 g) was added under argon and the reaction mixture was heated to reflux for 24 h. The mixture was filtered through celite and the filtrate was acidified with cone. HCl to pH~l. Concentration yielded a yellow oil which was purified by flash chromatography (MeOH/CH2Cl2 3:7+3.5% NH4 OH)to give 249 mg (51%) of 4-amino-l-methylpiperidine (57-MBT36B) as a white solid. Rf 0.13 (10% MeOH in CH2Cl2 +3.5% NH4OH). HPLC- MS (method B) showed MH+ =115. UV/MS(%)=-/100.
[0462] 4-Amino-l-methylpiperidine (57MBT36B) (26 mg, 0.231 mmol) was dissolved in methanol (1 mL) and 3-hydroxy-4-methylbenzaldehyde (32 mg, 0.231 mmol) and acetic acid (33μL) were added. The mixture was cooled to 0° C. NaBH3 CN (29 mg, 0.462 mmol) was added and the cooling bath was removed. After 3 h the reaction mixture was evaporated and flash chromatography (0-30% MeOH in CH2Cl2) gave 27 mg (50%) of N-((3-hydroxy-4-methylphenyl)methyl)-4-amino-l-methylpiperidine (57MBT44C) as a white solid. Rf =0.27 (10% MeOH in CH2Cl2 +3.5% NH4 OH). HPLC-MS (method A) showed MH+ =235. UV/MS(%)=99/99.
[0463] N-((3-hydroxy-4-methylphenyl)methyl)-4-amino- 1 -methylpiperidine (57MBT44C) (27 mg, 0.115 mmol) was dissolved in CH2Cl2 (2 mL). 4- Methoxyphenylacetyl chloride (17μL, 0.115 mmol) was added dropwise under argon. After 3 h, n-heptane (3 mL) was added and the mixture was evaporated. Flash chromatography (0-20% MeOH in CH2Cl2) gave 14 mg (32%) of the title compound as a white solid. Rf =0.32 (10% MeOH in CH2Cl2 +3.5% NH4 OH). HPLC-MS (method A) showed MH+ = 383. UV/MS(%)=99/96. 1H-NMR (400 MHz, CD3OD5 Rotamers 63:37): δ 7.28-6.55 (m, 7H), 4.48 (s, 2H), 4.37 and 3.95 (2m, IH)5 3.78 and 3.77 (2s5 3H), 3.06 and 2.89 (2br d, J=I 2 Hz5 2H)5 2.42 and 2.32 (2s, 3H)5 2.40 and 2.12 (2m, 2H), 2.18 and 2.12 (2s, 3H)5 1.86 and 1.83 (2m, 2H), 1.75 and 1.35 (2br d, J=12 Hz, 2H)
Example 89 - N-ff4-methylphenyl)methyl)-N-(l-methylpiperidin-4-yl)-2-('4- bromophenvDacet amide hvdrochloride(57MBT70-lD)
[0464] 4-Bromophenylacetic acid (54 mg, 0.252 mmol) was dissolved in CH2Cl2 (2 mL), and N-((4-methylphenyl)methyl)-4-amino-l -methylpiperidine (292 mg/mL stock solution in CH2Cl2, 171μL, 0.229 mmol) and polystyrene supported diisopropylethylamine (PS-DIEA with a loading of 3.57 mmol/g, 192 mg, 0.687 mmol) was added followed by bromo-tris-pyrrolidino-phosphonium hexafluorophosphate (PyBrOP, 160 mg/mL stock solution, 1 mL, 0.334 mmol). The reaction mixture was shaken for 1 h at r.t. and filtered onto a prewashed (methanol) ion exchange column (0.88 mmol/g, 1 g). The column was washed with methanol (8 X 4 mL) and the remaining product was eluted off the column with 10% NEL» OH in methanol (2 X 4 mL) and evaporated. The resulting oil was filtered through silica (H=4 cm, D=I cm)with methanol/CH2Cl2 1 :9 (20 mL), evaporated and subjected to a second ion exchange column (0.88 mmol/g. 1 g). The column was washed with methanol (8*4 mL) and the remaining product was eluted off the column with 10% NH4 OH in methanol (2*4 mL) and evaporated on rotavap and oil pump. The product was dissolved in CH2CI2 (0.5 mL) and HCl in diethylether (1.0 M, 0.1 mL, 0.1 mmol) was added. The solution was added to n-heptane (3 mL) and evaporation afforded 29 tng (25%) of the title compound as a white solid. Rf =0.31 (10% MeOH in CH2Cl2). HPLC-MS (method B) showed MH+ =416. UV/MS(%)=100/99.
Example 90 - N-C('4-methylphenyl)methyπ-N-('l-methylpiperidin-4-yl)-2-('4- iodophenvDaceta mide hvdrochloridef57MBT70-2D)
[0465] The title compound was prepared according to example MBT04. Yield: 33 mg (26%). Rf =0.31 (10% MeOH in CH2Cl2). HPLC-MS (method B)showed MH+ =463. UV/MS(%)=100/98.
Example 91 - N-(Y4-methylphenyl')methyl VN-(I -methylpiperidin-4-yl)-2-(4-(2- propyPphenyl acetamide hydrochloride f57MBT70-3D)
[0466] The title compound was prepared according to example MBT04. Yield: 36 mg (34%). Rf =0.31 (10% MeOH in CH2Cl2). HPLC-MS (method B) showed MH+ =379. UV/MS(%)=100/97.
Example 92 - N-r(4-methylphenyl)methyl)-N-d-methylpiperidin-4-yl)-2-f4- trifluoromethoxy phenvDacetamide hydrochloride(57MBT70-4D)
[0467] The title compound was prepared according to example MBT04. Yield: 35 mg (30%). Rf =0.27 (10% MeOH in CH2Cl2). HPLC-MS (method B) showed MH+ =421. UV/MS(%)=100/99.
Example 93 - N-ff4-methylphenyl)methyiyN-(l-methylpiperidin-4-viy2-(4- methylthiophenyl')acetamide hydrochloride(57MBT70-5D') [0468] The title compound was prepared according to example MBT04. Yield: 35 mg (33%). Rf =0.30 (10% MeOH in CH2Cl2). HPLC-MS (method B) showed MH+ =383. UV/MS(%)=100/99.
Example 94 - N-("(4-methylphenyl)methyl)-N-(l-methylpiperidin-4-yl')-2-(4-(N,N- dimethylami no)phenyl)acetamide hydrochloride^ 7MBT70-6D)
[0469] The title compound was prepared according to example MBT04.
[0470] Yield: 16 mg (15%). Rf =0.25 (10% MeOH in CH2Cl2). HPLC-MS (method A) showed MH+ =380. UV/MS(%)=100/100.
Example 95 - N-((4-methylphenyl)methyl')-N-(l-methylpiperidin-4-vπ-2-('4- nitrophenvDacet amide hvdrochloride^MBTyO^D')
[0471] The title compound was prepared according to example MBT04. Yield: 28 mg (27%). Rf =0.27 (10% MeOH in CH2Cl2). HPLC-MS (method B) showed MH+ =382. UV/MS(%)=100/100.
Example 96 - N-((4-methylphenyl)methyl)-N-ri-rhethylpiperidin-4-yl)-2-r4-methoxy-3- methyl phenvDacetamide hvdrochloride(57MBT70-8D>)
[0472] The title compound was prepared according to example MBT04. Yield: 34 mg (32%). Rf =0.30 (10% MeOH in CH2Cl2). HPLC-MS (method B) showed MH+ =381. UV/MS(%)=100/99.
Example 97 - N-f(4-memylphenvDmethyl>N-(l-methylpiperidin-4-yl)-2-(4- pyridvDacetamid e hydrochloride(57MBT70-9F)
[0473] The title compound was prepared according to example MBT04. Yield: 18 mg (17%). Rf =0.09 (10% MeOH in CH2Cl2). HPLC-MS (method A) showed MH+=338. UV/MS(%)=100/100.
Example 98 - N-((4-methylphenyl)methylVN-π-methylpiperidin-4-yl)-2-(4- methylphenvDace tamide hydrochloridef57MBT62B)
[0474] The title compound was prepared according to example MBT04. Yield: 10 mg (35%). Rf =(10% MeOH in CH2Cl2). HPLC-MS (method A) showed MH+=SSl . UV/MS(%)=100/100. Example 99 - N-((4-πiydroxymethyl)phenvπmethylVN-(l-methylpiperidin-4-yl)-2-('4- methoxy phenvDacetamide hydrochloride(57MBT72D)
[0475] To a stirred suspension of LiAlHU (285 mg, 7.52 mmol) in diethylether (10 mL)at 0° C. was added a solution of 4-cyanobenzyl alcohol (0.5 g, 3.76 mmol) in diethylether (5 mL) over 15 min. The grey reaction mixture was heated to reflux for 3 h. After cooling to r.t, the mixture was treated successively with water (1 mL), 2M NaOH (2 mL) and water (2 mL) under vigorous stirring. The resulting white slurry was filtered and washed with CH2Cl2 (20 mL). Extraction with additional CH2Cl2 (20 mL) and n- butanol (20 mL) and evaporation yielded an oil, which upon flash chromatography (0- 15% MeOH in CH2Cl2) gave 152 mg (29%)of 4-(aminomethyl)benzylalcohol (57MBT52B) as a white solid. Rf=0.51 (30% MeOH in CH2Cl2 +3.5% NH4 OH).
[0476] l-Methyl-4-piperidone (84μL, 0.73 mmol) was dissolved in methanol (5 mL) and 4-(aminomethyl)benzylalcohol (57MBT52B) (100 mg, 0.73 mmol) was added followed by acetic acid (125μL). NaBH3 CN (92 mg, 1.46 mmol) was added and the mixture was stirred for 3 h. The reaction mixture was evaporated and 2M NaOH (5 mL) was added. Extraction with CH2Cl2 (4 X 5 mL), drying with Na2SO4 and evaporation gave 152 mg (87%) of N-((4-(hydroxymethyl)phenyl)methyl)-4-amino-l- methylpiperidine (57MBT56D) as a white solid. HPLC-MS (method B) showed MH+ =235. UV/MS(%)=100/100.
[0477] N-((4-(Hydroxymethyl)phenyl)methyl)-4-amino- 1 -methylpiperidine (57MBT56D) (20 mg, 0.0853 mmol) was dissolved in CH2Cl2 (2 mL) and 4- methoxyphenylacetyl chloride (26μL, 0.171 mmol) was added dropwise. The reaction mixture was stirred for 1 h and water (500μL) was added followed by evaporation. A solution of sodium (5 mg, 0.179 mmol) in methanol (2 mL) was added. After stirring for 4 h, the solution was transferred to a prewashed (methanol) ion exchange column (0.88 mmol/g, 1 g) and washed with methanol (4 X 4 mL). The remaining product was eluted off the column with 10% NH4 OH in methanol (2 X 4 mL) and evaporated. The resulting oil was filtered through silica (H=4 cm, D=I cm) with methanol/CH2Cl2 2:8 (20 mL), evaporated and subjected to a second ion exchange column (0.88 mmol/g, 1 g). The column was washed with methanol (8 X 4 mL) and the remaining product was eluted off the column with 10% NH4 OH in methanol (2 X 4 mL) and evaporated on rotavap and oilpump. The product was dissolved in CH2Cl2 (0.5 mL) and HCl in diethylether (1.0 M, 0.1 mL, 0.1 mmol) was added. The solution was added to n-heptane (3 mL) and evaporation afforded 14 mg (39%) of the title compound as a white solid. Rf=0.16 (10% MeOH in CH2Cl2). HPLC-MS (method B) showed MH+ =383. UV/MS(%)=100/96.
Example 100 - 2-(4-ChlorophenylVN-('4-methylbenzyl')-N-(l-isopropylpiperidin-4- vnacetamid e (47AKU-7)
1 -Trifluoroacetyl-4-piperidone (47 AKU-2)
[0478] 4-Piperidone hydrochloride monohydrate (3.85 g, 25 mmol) and Triethylamine (10.5 ml, 75 mmol) were partly dissolved in 100 ml of dichloromethane and stirred for 10 min. Reaction mixture was then cooled on ice-bath and trifluoroacetic anhydride (7.2 ml, 50 mmol) was slowly added over 10 min. Ice-bath was removed and mixture was stirred overnight. Additional trifluoroacetic anhydride (2 ml) was added and the mixture was stirred for 1 hr. Water (200 ml) was added. Phases were separated and aq. phase was re-extracted with dichloromethane. Combined organic phases were washed with brine, dried over MgSO4 and concentrated (40° C.) giving 4.97 g (100%) 47AKU-2 as yellow crystals. TLC (5% methanol in dichloromethane): Rf =0.8. 1H-NMR (400 MHz5 CDCl3): δ 3.87-3.99 (4H, m); 2.54-2.61 (4H, m). 13C-NMR (CDCl3): δ 204.7, 118.0, 1 15.1, 44.2, 42.8, 41.2, 40.5.
4-(4-MethylbenzylaminoVl -trifluoroacetyl-piperidine (47AKU-3)
[0479] 47AKU-2 (4.97 g, 25 mmol) was dissolved in 100 ml methanol and 4- methylbenzyl-amine (3.2 ml, 25 mmol) was added. Mixture was stirred and acetic acid (~2 ml) was added until pH~5. NaCNBH3 (3.15 g, 50 mmol) was slowly added. After magnetic stirring for 20 hrs the methanol was partly removed on the rotary evaporator (40° C). Dichloromethane, 2M NaOH and water were added until pH~10. Phases were separated and aq. phase was then re-extracted twice with dichloromethane. Combined organic phases were washed with brine and dried over MgSO4. Concentration(40° C.) yielded 6.94 g (92%) 47AKU-3. TLC (10% methanol in dichloromethane): Rf =0.6. HPLC-MS (Method A): M+ =301.0 (UV/MS(%)=94/100).
2-(4-ChlorophenylVN-(4-methylbenzyl)-N-(l-trifluoroacetylρiperidin-4-yl')ac etamide f47AKU-4*>
[0480] 47AKU-3 (3.01 g, 10 mmol) in 25 ml of dichloromethane was placed in a 100 ml flask. Triethylamine (1.4 ml, 10 mmol) was added and the mixture was cooled on an ice-bath and stirred for 10 min. 4-Chlorophenylacetyl chloride (1.90 g, 10 mmol) was dissolved in 10 ml dichloromethane and added slowly to the ice-cold mixture. After 15 min. the ice-bath was removed and the mixture was left for 1 hr. Precipitation was observed. The reaction mixture was then concentrated at aspirator pressure(40° C). The crude product was purified by flash chromatography (0-50% ethylacetate in heptane) yielding 2.38 g (53%/) 47AKU-4. TLC (100% dichloromethane): Rf =0.6. HPLC-MS (Method A): M+ =453.0 (UV/MS(%)=89/84).
2-(4-ChloroDhenylVN-f4-methylbenzvtVN-rpiperidin-4-vnacetamide (47AKU-6')
[0481] 47AKU-4 (2.38 g; ~5 mmol) was dissolved in 50 ml of methanol. K2CO3 (3.5 g; 25 mmol) was added in one portion. After magnetic stirring for 20 hrs, additional K2CO3 (1 g) was added. After 4 hrs magnetic stirring methanol was partly removed by evaporation(40° C). Ethyl acetate (100 ml) and water (100 ml) were added. The phases were separated and the aq. phase was then re-extracted with ethylacetate. The combined organic phases were dried over MgSO4 and concentrated (40° C.) giving 1.95 g (100%) 47AKU-6. TLC (20% methanol in dichloromethane): Rf =0.3. HPLC-MS (Method A): M+ =357.1 (UV/MS(%)=84/95).
2-(4-Chlorophenyl)-N-(4-methylbenzyl)-N-( 1 -isopropylpiperidin-4-yl)-acetami de (47AKU-7)
[0482] 47AKU-6 (358 mg, 1.0 mmol) was dissolved in 20 ml of acetonitrile. Triethylamine (1.4 ml, 10 mmol) was added and mixture was stirred for 10 min. Isopropyl bromide (370 mg, 3.0 mmol) was dissolved in 5 ml of acetonitrile and added to the reaction mixture which was stirred at room temp, for 20 hrs and then heated to 60° C. for 4 hrs. After cooling, ethylacetate (25 ml) and water (25 ml) were added. The phases were separated and the aq. phase was then re-extracted with ethylacetate. The combined organic phases were washed with brine, dried over MgSO4 and concentrated (40° C.) giving 362 mg of crude product. Purification by flash chromatography (0-10% methanol in dichloromethane) and HCl-precipitation from 2M HCl/diethyl ether in dichloromethane/heptane gave 76 mg (18%) 47AKU-7. TLC (10% methanol in dichloromethane): Rf =0.4. Mp=223-224° C HPLC-MS (Method A): M+ =399.1 (UV/MS(%)=100/99). 1H-NMR (400 MHz, CDCl3): δ 7.03-7.29 (8H, m); 4.86 (IH, m); 4.61 (2H, m); 3.58 (2H, m); 3.37 (3H, m); 2.82 (2H5 m); 2.64 (2H, m); 2.34 (3H, s); 1.80 (2H, m); 1.39 (6H, d). 13C-NMR (CDCl3): δ 172.4 137.4, 134.8, 133.3, 133.1, 130.4, 129.9, 129.0, 125.8, 58.0, 49.5, 48.2, 46.6, 40.4, 26.0, 21.2, 17.0. Example 101 - 2-(4-Chlorophenyl)-N-(4-methylbenzylVN-(l-ethylpiperidin-4- vnacetamide (47AKU-12)
[0483J 47AKU-6 (358 mg, 1.0 mmol) was dissolved in 20 ml of acetonitrile. Triethylamine (1.4 ml, 10 mmol) was added and the mixture was stirred for 10 min. Ethyl bromide (370μl, 5.0 mmol) was added. The mixture was then heated to 50° C. and stirred overnight. After cooling, water (25 ml) and ethylacetate (25 ml) were added. The phases were separated and the aq. phase was re-extracted with ethylacetate. The combined organic phases were washed with brine and dried over MgSO4. Evaporation(40° C.) yielded 406 mg of crude product. Purification by ion exchange chromatography (washout with 10% aq. NH4OH (25%) in methanol) gave 166 mg (43%) 47AKU-12. The HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.5. HPLC-MS (Method A): M+ =385.1 (UV/MS(%)= 100/99). 1H-NMR (400 MHz, CDCl3, rotamers): δ 7.02-7.34 (8H, m); 4.62 (IH, m); 4.46 and 4.53 (2H, 2s); 3.81 (IH, s); 3.55 (2H, s); 2.92 (2H, m); 2.34 (3H, s); 2.29 (IH, s); 1.98 (2H, m); 1.52-1.84 (4H5 m); 1.03 (3H, t). 13C-NMR (CDCl3): δ 171.7, 137.2, 135.4, 133.9, 132.8, 130.4, 129.7, 128.9, 125.8, 52.8, 52.4, 46.5, 40.8, 31.2, 29.8, 21.2, 12.4.
Example 102 - 2-Phenyl-N-f4-methylbenzyl)-N-d-methylpiperidin-4-yl)-acetamide (47AKU-13Ϊ
[0484] 47AKU-5 (218 mg, 1.0 mmol) was dissolved in 2 ml of dichloromethane in a 50 ml flask. Phenylacetyl chloride (134μl, 1.0 mmol) was added. After 3 hrs stirring at room temp, mixture was concentrated on Rotavapor (40° C). Crude product was purified by ion exchange chromatography (washout with 10% aq. NH4OH (25%) in methanol) and flash chromatography (0-10% methanol in dichloromethane) giving 48 mg (14%) 47AKU-13. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ =337.1 (UV/MS(%)=98/98). IH-NMR (400 MHz, CDC13, rotamers): δ 7.01-7.40 (9H, m); 4.63 (IH, m); 4.53 and 4.45 (2H, 2s); 3.85 and 3.61 (2H, 2s); 2.86 and 2.77 (2H, 2m); 2.35 and 2.29 (3H, 2s); 2.25 and 2.20 (3H, 2s); 209 (2H, m); 1.61-1.86 (4H, m). 13C-NMR (CDC13): δ 72.2, 137.1, 135.5, 129.7, 128.9, 128.8, 127.2, 126.9, 125.8, 55.3, 5L6, 46.6, 46.1, 41.6, 29.5, 21.2. Example 103 - 2-f4-ChlorophenylVN-(4-methylbenzylVN-('l-methylpiperidin-4-yl)- acetamide f47AKU-8')
4-(4-MethylbenzylaminoV 1 -methyl-piperidine (47 AKU-5")
[0485] l-Methyl-4-piperidone (1.13 g, 10 mmol) was dissolved in 20 ml of methanol and added to a 100 ml flask. 4-Methylbenzylamine (1.21 g, 10 mmol) in 10 ml of methanol was added. Acetic acid (~1.5 ml) was added until pH~5. NaCNBEk (1.26 g, 20 mmol) was slowly added. After 20 hrs magnetic stirring methanol was partly removed on Rotavapor (40° C). Dichloromethane, water and 2M NaOH were added until pH~10. The phases were separated and aq. phase was extracted twice with dichloromethane. The combined organic phases were washed with brine and dried over MgSO4. Concentration on Rotavapor (40° C.) yielded 2.06 g crude (93%) 47 AKU-5. TLC (20% methanol in dichloromethane): Rf=0.3. HPLC-MS (Method A): M+ =219.1 (UV/MS(%)=89/98).
2-(4-Chlorophenyl)-N-r4-methylbenzyl)-N-π-methylpiperidin-4-yl)-acetamide (47AKU-8>
[0486] 47 AKU-5 (437 mg, 2.0 mmol) was dissolved in 10 ml of dichloromethane in a 50 ml flask. Triethylamine (280μl, 2.0 mmol) was added and the mixture was cooled to 0° C. on an ice bath and stirred for 10 min. 4-Chlorophenylacetyl chloride (380 mg, 2.0 mmol) was dissolved in 10 ml of dichloromethane and added to the cooled mixture. After 2 hrs stirring at room temp, additional dichloromethane (10 ml) and water (20 ml) were added. The phases were separated and the aq. phase was re-extracted with dichloromethane. The combined organic phases were dried over MgSO4 and concentrated on the Rotavapor (40° C.) giving 755 mg of crude product. Purification by flash chromatography (0-10% methanol in dichloromethane)gave 485 mg (65%) product. Further purification by ion exchange chromatography (washout with 10% aq. NH4OH (25%) in methanol) gave 239 mg (32%) 47AKU-8. The HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.4. Mρ=217-219° C. HPLC-MS (Method A): M+ =371.1 (UV/MS(%)=99/99). 1H- NMR (400 MHz, CD3OD): δ 7.05-7.39 (8H5 m); 4.80 (3H, s); 4.62+4.56 (2H, 2s); 4.35 (IH, m); 4.00 (IH3 s); 3.71 (IH, s); 3.46 (2H, m); 3.06 (2H, m); 2.80 (3H5 s); 2.32+2.27 (3H5 2s); 2.19 (lH,m). 13C-NMR (CD3OD): δ 173.0, 137.5, 134.5, 133.9, 132.6, 130.6, 129.5, 128.5, 126.2, 54.0, 51.4, 42.6, 40.2, 31.8, 26.6, 19.9. Example 104 - 2-f4-ChlorophenylVN-(4-methylberizΛd>N-π-cyclopentylpiperidin-4-yl)- aceta mide f 47 AKU-I 1Λ
[04871 47AKU-6 (358 mg, 1.0 mmol) was dissolved in 20 ml of acetonitrile. Triethylamine (1.4 ml, 10 mmol) was added and mixture was stirred for 10 min. Cyclopentylbromide (540μl, 5.0 mmol) was added and the mixture was stirred at room temp. After 20 hrs the mixture was heated to 50° C. for an additional 24 hrs. The reaction mixture was then cooled and water (25 ml) and ethylacetate (25 ml) were added. The phases were separated and the aq. phase was re-extracted with ethylacetate. The combined organic phases were washed with brine and dried over MgSO4. Concentration on Rotavapor (45° C.) yielded 426 mg of crude product. Purification by ion exchange chromatography (washout with 10% aq. NH4 OH (25%) in methanol) and flash chromatography (0-10% methanol in dichloromethane)gave 76 mg (18%) 47AKU-11. The HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.5. HPLC-MS (Method A): M+ =425.1 (U V/MS(%)=100/97). 1H-NMR (400 MHz5 CDCl3, rotamers): δ 7.01-7.34 (8H, m); 4.67 (IH, m); 4.49 and 4.52 (2H3 2s); 3.54 (2H, s); 3.15 and 3.02 (2H, 2m); 2.64 (IH, m); 2.27 and 2.34 (3H, 2s); 2.20 (IH, m); 1.85 (4H, m); 1.69 (4H3 m); 1.53 (4H, m); 1.37 (IH3 m). 13C-NMR (CDCl3): δ 171.9, 137.2, 135.2, 133.8, 132.9, 130.4, 129.7, 128.9, 125.8, 67.7, 52.4, 52.1, 46.5, 40.7, 30.2, 28.8, 24.3, 21.2.
Example 105 - 2-(4-Fluoro phenyl VN-(4-methylbenzyl>N-π-methylpiperidin-4-yl*)- acetamide f47AKU-14~)
[0488] 47AKU-5 (218 mg, 1.0 mmol) was dissolved in 3 ml of dichloromethane in a 50 ml flask. 4-Fluorophenylacetyl chloride (150μl, 1.1 mmol) was added. After 4 hrs stirring at room temp, the mixture was concentrated on Rotavapor (40° C). The crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 243 mg (68%) 47AKU-14. The HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf=0.5. HPLC-MS (Method A): M+ =355.1 (UV/MS(%)=100/100). 1H-NMR (400 MHz, CDCl3): δ 6.92-7.33 (8H, m); 4.73 (IH, m); 4.52 (2H, s); 3.56 (2H5 2s); 3.44 (5H, m); 3.25 (2H, m); 2.52-2.67 (4H, m); 2.33 (3H, s). 13C-NMR (CDCl3): δ 172.5, 163.3, 160.9, 139.5, 134.8, 130.6, 129.8, 125.8, 115.8, 54.6, 50.8, 49.9, 46.7, 40.4. 27.2, 21.2. Example 106 - 2-(4-ChlorophenylVN-r4-methylben2ylVN-π-2-hvdroxyethylVpiperidin- 4-ylV acetamide (47AKU-IS^
[0489] 47AKU-6-2 (358 mg, 1.0 mmol) was dissolved in 10 ml of acetonitrile in 50 ml flask. Triethylamine (1.4 ml, 10 mmol) was added and mixture was stirred for 10 min.
[0490] 2-Bromoethanol (215μl, 3.0 mmol) was added. Reaction mixture was then heated to 60° C. and stirred overnight. After cooling ethylacetate (25 ml) and water (25 ml) were added. Phases were separated and aq. phase was re-extracted with ethylacetate. Combined organic phases were washed with brine, dried over MgSCU and concentrated on Rotavapor (40° C.) giving 406 mg crude product. Purification by flash chromatography (0-10% methanol in dichloromethane) afforded 253 mg (63%) 47 AKU- 18. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ =401.1 (UV/MS(%)=100/100). 1H7NMR (400 MHz3 CDCl3, rotamers): δ 7.04-7.34 (8H, m); 4.60 (IH5 m); 4.52 and 4.45 (2H5 2s); 3.55 (4H, m); 3.03 (IH5 bs); 2.92 (2H, m); 2.52 (2H, m); 2.36 and 2.31 (3H5 2s); 2.19 (2H, m); 1.66 (4H, m). ). 13C-NMR (CDCl3): δ 171.7, 137.3, 135.2, 133.8, 132.9, 130.4, 129.8, 128.9, 125.8, 59.4, 58.1, 53.1, 52.3, 46.8, 40.8, 29.7, 21.2.
Example 107 - 2-(4-Chlorophenyl)-N-(4-methylbenzyl)-N-(l-cvclobutylpiperidin-4-yl)- acetam ide (47AKU-19)
1 -Cvclobutyl-4-ρiperidone f 47 AKU-15>
[0491] Partly dissolved quartenary salt (1.23 g, 3.7 mmol) (prepared according to the procedure outlined in the synthesis of 47AKU-47) was slowly added to a refiuxing solution of Cyclobutylamine (178 mg, 2.5 mmol) and Potassium carbonate (48 mg, 0.34 mmol) in ethanol. The mixture was refluxed for 1.5 hrs. After cooling to room temp, water (10 ml) and dichloromethane (25 ml) were added. Phases were separated and aq. phase was re-extracted with dichloromethane. Combined organic phases were dried over MgSO4 and concentrated on Rotavapor (40° C.) giving 419 mg crude 47AKU-15. TLC (10% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ =154.1 (MS(%)=75).
4-f4-Methylbenzylamino)-l -cvclobutyl-piperidine (47AKU- 16) [0492J 4-Methylbenzylamine (215 mg, 1.8 mmol) was dissolved in 5 ml methanol and placed in 50 ml flask. 47 AKU-15 (270 mg, 1.8 mmol) in 5 ml methanol was added. Acetic acid (0.3 ml) was added until pH~5. NaCNBH3 (226 mg, 3.6 mmol) was slowly added. Gas evolution observed. After 24 hrs magnetic stirring dichloromethane, 2M NaOH and water were added until pH~10. Phases were separated and aq. phase was then re-extracted with dichloromethane. Combined organic phases were dried over MgSO4 and concentrated on Rotavapor (40° C.) yielding 419 mg crude 47AKU-16. TLC (10% methanol in dichloromethane): Rf =0.3. HPLC-MS (Method A): M+ =259.1 (UV/MS(%)=44/87).
2-f4-Chlorophenyl)-N-(4-methylbenzyl)-N-d-cvclobutylpiperidin-4-yl')-acetam ide C47AKU-19')
[04931 47AKU-16 (209 mg, 0.8 mmol) was placed in 50 ml flask and 5 ml dichloromethane was added. 4-Chlorophenylacetyl chloride (171 mg, 0.9 mmol) in 5 ml dichloromethane was added. After 5 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 101 mg (31%) product. Further purification by ion exchange chromatography (washout with 10% aq. NH4 OH (25%) in methanol) gave 55 mg (17%) 47AKU-19. Oxalate-salt was prepared from Oxalic acid (1.1 eq) in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.6. HPLC-MS (Method B): M+ =411.2 (UV/MS(%)=91/86). 1H-NMR (400 MHz, CDCl3, rotamers): δ 7.33-7.01 (8H, m); 4.62 (IH, m); 4.52 and 4.46 (2H, 2s); 3.80 (IH, s); 3.45 and 3.54 (2H, 2s); 2.86 (2H, m); 2.66 (2H, m); 2.28 and 2.34 (3H, 2s); 1.98 (2H, m); 1.80 (2H, m); 1.70- 1.52 (6H, m). 13C-NMR (CDCl3): δ 171.7, 137.2, 135.4, 133.9, 132.9, 130.4, 129.7, 128.9, 125.7, 60.4, 52.3, 49.4, 46.5, 40.7, 29.4, 27.6, 21.2, 14.2.
Example 108 - 2-C4-Methoxyphenyl*)-N-(4-methylbenzyl')-N-fl-cvclobutylpiperidin-4- vDacetam ide (47AKU-20)
[0494] 47AKU-16 (209 mg, 0.8 mmol) was placed in 50 ml flask and 5 ml dichloromethane was added. 4-Methoxyphenylacetyl chloride (167 mg, 0.9 mmol) in 5 ml dichloromethane was added. After 5 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 72 mg (22%) product. Further purification by ion exchange chromatography (washout with 10% aq. NH4 OH (25%) in methanol) gave 67 mg (20%) 47AKU-20. Oxalate-salt was prepared from Oxalic acid (1.1 eq) in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf=0.6. HPLC-MS (Method B): M+ =407.3 (UV/MS(%)=93/77). 1H-NMR (400 MHz, CDCl3, rotamers): δ 7.26-6.79 (8H5 m); 4.62 (IH, m); 4.52 and 4.45 (2H5 2s); 3.79 (IH, m); 3.77 (3H, s); 3.52 and 3.45 (2H, 2s); 2.84 (2H3 m); 2.66 (2H5 m); 2.34 and 2.28 (3H, 2s); 1.98 (2H, m); 1.81 (2H5 m); 1.72-1.51 (6H,m). 13C-NMR (CDCl3): δ 172.5, 158.7, 137.0, 135.7, 130.4, 129.8, 127.4, 125.8, 114.3, 60.4, 55.5, 52.1, 49.4, 46.4, 40.6, 29.4, 27.6, 21.2, 14.2.
Example 109 - (47AKU-2U 2-f4-MethoxyphenylVN-(4-methylbenzylVN-r8-methyl-8- aza-bicvclof3.2.11oct-3-ylVacetamide T47AKU-21 )
4-f4-MethylbenzylaminoVtropane (47 AKU-17)
[0495] 4-Methylbenzylamine (607 mg, 5.0 mmol) was dissolved in 10 ml methanol and placed in 100 ml flask. Tropinone (697 mg, 5.0 mmol) in 10 ml methanol was added. Acetic acid (0.75 ml) was added until pH~5. NaCNBH3 (628 mg, 10 mmol) was slowly added. Gas evolution observed. After 20 hrs magnetic stirring dichloromethane, 2M NaOH and water were added until pH~10. Phases were separated and aq. phase was then re-extracted with dichloromethane. Combined organic phases were dried over MgSO4. Concentration on Rotavapor (40° C.) yielded 1.14 g crude 47AKU-17. TLC (10% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ -245.2 (UV/MS(%)=65/96).
2-^4-Methoxyphenvn-N-f4-methylbenzylVN-r8-methyl-8-aza-bicvclo^3■2■noct-3- ylVacetamide (47AKU-2H
[0496] 47AKU-17 (244 mg, 1.0 mmol) was placed in 50 ml flask and 5 ml dichloromethane was added. 4-Methoxyphenylacetyl chloride (203 mg, 1.1 mmol) in 10 ml dichloromethane was added. After 3 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by ion exchange chromatography (washout with 10% aq. NH4 OH (25%) in methanol) and flash chromatography (0-10% methanol in dichloromethane) giving 202 mg (51%) 47AKU-21. Oxalate-salt was prepared from Oxalic acid (1.1 eq) in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf 0.4. HPLC-MS (Method B): M+ =393.3 (UV/MS(%)=94/92). 1H-NMR (400 MHz, CDCl3, isomers): δ 7.02-7.17 (6H, m); 6.78- 6.87 (2H, m); 4.74 (IH, s); 4.44 (IH, s); 3.78 and 3.77 (3H, 2s); 3.68 (IH, m); 3.66 and 3.55 (3H, 2s); 2.65 (2H, m); 2.56 (2H5 m); 2.32 (3H, s); 2.12-2.26 (6H, m); 2.05 (2H, m). 13C-NMR (CDCl3): δ 173.2, 171.4, 158.8, 137.1, 129.7, 127.6, 126.9, 126.0, 114.4, 63.4, 60.9, 55.5, 54.6, 47.5, 41.5, 40.4, 32.8, 31.1, 27.5, 24.9, 21.2.
Example UO - N-(4-Methylben2yl")-N-fl-methylpiperidin-4-ylVN'-benzyl-carbainide (47AKU-22)
[0497] 47AKU-5 (219 mg, 1.0 mmol) was dissolved in 5 ml dichloromethane and placed in 50 ml flask. Benzylisocyanate (160 mg, 1.2 mmol) in 5 ml dichloromethane was added. After 16 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 236 mg (67%) 47AKU-22. Oxalate-salt was prepared from Oxalic acid (1.1 eq) in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.5. HPLC-MS (Method B): M+ =352.3 (UV/MS(%)=100/100). 1H-NMR (400 MHz, CDCl3): δ 7.26-7.02 (9H, m); 4.61 (IH, m); 4.41 (IH, m); 4.33 (4H, rn); 2.87 (2H, m); 2.32 (3H, s); 2.25 (3H, s); 2.09 (2H, m); 1.79-1.62 (4H, m). 13C-NMR (CDCl3): δ 158.6, 139.7, 137.3, 135.4, 129.8, 128.6, 127.4, 127.2, 126.2, 55.5, 52.2, 46.2, 45.8, 45.0, 30.2, 21.2.
Example 111 - N-f4-MethylbenzylVN-ri-methylpiρeridin-4-yl')-N'-phenyl carbamide (47AKU-24)
[0498] 47AKU-5 (219 mg, 1.0 mmol) was dissolved in 5 ml dichloromethane and placed in 50 ml flask. Phenylisocyanate (143 mg, 1.2 mmol) in 5 ml dichloromethane was added. After 4 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 181 mg (54%) 47AKU-24. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ =338.3 (UV/MS(%)=100/100). 1H-NMR (400 MHz5 CDCl3): δ 7.12-7.24 (8H, m); 6.93-6.98 (IH, m); 6.26 (IH, s); 4.45 (3H, s); 2.90 (2H, d); 2.36 (3H, s); 2.28 (3H, s); 2.12 (2H, m); 1.69-1.85 (4H, m). 13C- NMR (CDCl3): δ 156.1, 139.3, 137.8, 134.9, 130.1, 128.9, 126.3, 123.1, 119.9, 55.5, 52.3, 46.3, 46.2, 30.3, 21.3. Example 112 - N-Phenethyl-N-π-methylpiperidin-4-vn-N'-benzyl-carbamide C47AKU- 25}
[0499] 4-(2-Phenylethyl)amino-l-methylpiperidine (110 mg, 0.5 mmol) was dissolved in 5 ml dichloromethane and placed in 50 ml flask. Benzylisocyanate (80 mg, 0.6 mmol) in 5 ml dichloromethane was added. After 20 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 164 mg (84%) 47AKU-25. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ =352.3 (UV/MS(%)=100/100). 1H-NMR (400 MHz, CDCl3): δ 7.34-7.09 (1OH, m); 4.52 (IH, m); 4.35 (2H5 d); 4.08 (IH, m); 3.33 (2H, t); 2.92 (2H, m); 2.82 (2H, t); 2.28 (3H5 s); 2.07 (2H5 m); 1.84-1.66 (4H5 m). 13C-NMR (CDCl3): δ 157.9, 139.8, 139.1, 129.0, 128.9, 128.8, 127.8, 127.4, 126.9, 55.7, 52.8, 46.2, 45.3, 44.8, 37.5, 30.6.
Example 113 - 2-Phenyl-N-f4-methoxybenzylVN-π-methyIpiperidin-4-ylVacetamide (47AKU-26a)
[0500] 50ELH-18 (118 mg, 0.5 mmol) was dissolved in 5 ml dichloromethane in 50 ml flask.
[0501] 4-Fluorophenylacetyl chloride (104 mg, 0.6 mmol) was added. After 20 hrs stirring at room temp, mixture was concentrated on Rotavapor (400C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 87 mg (49%) 47AKU-26a. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. HPLC-MS (Method A): M+ =353.1 (UV/MS(%)=96/88).
Example 114 - 2-(4-Trifluoromethylρhenyl)-N-('4-methoxybenzyl)-N-(l-methylpiperidin- 4-ylV acetamide (47AKU-26K)
[0502] 50ELH-18 (1 18 mg, 0.5 mmol) was dissolved in 5 ml dichloromethane in 50 ml flask.
[0503] 4-Trifluoromethylphenylacetyl chloride (134 mg, 0.6 mmol) was added. After 20 hrs stirring at room temp, mixture was concentrated on Rotavapor (40° C). Crude product, was purified by flash chromatography (0-10% methanol in dichloromethane) giving 81 mg (39%) 47AKU-26b. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. HPLC-MS (Method A): M+ =421.1 (UV/MS(%)=90/l 00).
Example 115 - 2-(4-F.uorophenylVN-(4-methoxybenzyl)-N-(l-methylpiperidin-4-yl)- acetamide C47AKU-26(Λ
[0504] 50ELH-18 (118 mg, 0.5 mmol) was dissolved in 5 ml dichloromethane in 50 ml flask.
[0505] 4-Fluorophenylacetyl chloride (104 mg, 0.6 mmol) was added. After 20 hrs stirring at room temp, mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 68 mg (37%) 47AKU-26C. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. HPLC-MS (Method A): M+ =371.1 (UV/MS(%)=100/97).
Example 1 16 - 2-(4-Methoxyphenvπ-N-(4-methoxybenzvπ-N-π-methylpiperidin-4-yl)- acetamid e (47AKU-26d)
[0506] 50ELH-18 (118 mg, 0.5 mmol) was dissolved in 5 ml dichloromethane in 50 ml flask.
[0507] 4-Methoxyphenylacetyl chloride (111 mg, 0.6 mmol) was added. After 20 hrs stirring at room temp, mixture was concentrated on Rotavapor (40° C)- Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 77 mg (40%) 47AKU-26d. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. HPLC-MS (Method A): M+ =383.1 (UV/MS(%)=100/100).
Example 117 - 2-(4-Methylphenyl)-N-r4-chlorobenzvD-N-(l-methylpiperidin-4-yl)- acetamide C47AKU-28'>
4-f4-ChlorobenzylamineH -methyl-piperidine C47AKU-27>)
[0508] 1 -Methyl -4-piperidone (566 mg, 5.0 mmol) was dissolved in 10 ml methanol and placed in 100 ml flask. 4-Chlorobenzylamine (708 mg, 5.0 mmol) was added. Mixture was stirred and Acetic acid (-0.75 ml) was added until pH~5. NaCNBH3 (628 mg, 10 mmol) was slowly added. Gas evolution observed. After magnetic stirring for 16 hrs methanol was partly removed on Rotavapor (40° C). Dichloromethane, 2M NaOH and water were added until pH~10. Phases were separated and aq. phase was then re- extracted with dichloromethane. Combined organic phases were dried over MgSO4. Concentration on Rotavapor (40° C.) yielded 1.14 g crude 47AKU-27. TLC (10% methanol in dichloromethane): Rf=0.3. HPLC-MS (Method A): M+ =239.1 (MS(%)=96).
2-r4-MethvIphenylVN-f4-chlorobenzyl)-N-π-methylpiperidin-4-ylVacetamide (47AKU-28^
[0509] p-Tolylacetic acid (1.50 g) was dissolved in 10 ml thionylchloride and placed in 50 ml flask. Mixture was heated to reflux for 2 hrs and then concentrated on Rotavapor (40° C).
[0510] p-Tolylacetic chloride (202 mg, 1.2 mmol) in 5 ml dichloromethane was added to 47AKU-27 (239 mg, 1.0 mmol) in 5 ml dichloromethane. After 4 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 104 mg (28%) 47AKU-28. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.5. HPLC-MS (Method A): M+ =371.1 (UV/MS(%)=100/90). 1H-NMR (400 MHz5 CDCl3, rotamers): δ 7.34-6.99 (8H5 m); 4.57 (IH5 m); 4.50 and 4.44 (2H, 2s); 3.80 (IH5 s); 3.55 (IH5 s); 2.96 and 2.82 (2H5 2m); 2.34 (IH5 m); 2.32 (3H, s); 2.24 and 2.15 (3H5 2s); 1.91 (IH5 m); 1.81-1.59 (4H5 m). 13C-NMR (CDCl3): δ 172.5, 138.2, 136.8, 133.4. 131.8, 129.7, 129.2, 128.6, 127.4, 54.9, 51.3, 46.7, 41.3, 30.6, 28.6, 21.2.
Example 118 - 2-f4-HydroxyphenylVN-('4-methylbenzylVN-(l-methylpiperidin-4-vπ- acetamide f47AKU-29^
[0511] 42ELH-77 (41 mg, 0.1 mmol) was dissolved in 1 ml dry dichloromethane and placed in oven-dried 10 ml flask. Mixture was cooled to -78° C on a dry-ice/isopropanol bath. Borontribromide (1.0 M in dichloromethane, 150μl, 0.15 mmol) was slowly added at -78° C Ice-bath was removed and mixture was left at room temp, for 2 hrs. Water (3 ml) and saturated NaCl (aq.) were added and aq. phase was extracted with dichloromethane, ethylacetate and n-butanol. Combined organic phases were dried over MgSO4 and concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-20% methanol in dichloromethane) giving 22 mg (63%) 47AKU-29. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.3. HPLC-MS (Method A): M+ =353.2 (UV/MS(%)=100/100). 1H-NMR (400 MHz, CDCl3, rotamers): δ 7.07-6.60 (8H, m); 4.48 (IH. m); 4.39 (2H, s); 3.76 and 3.66 (4H, 2bs); 3.41 (2H, s); 3.08 (2H5 m); 2.49 (IH5 m); 2.42 (2H5 bs); 2.22 and 2.16 (3H5 2s); 1.96-1.82 (2H5 m); 1.66-1.56 (IH5 m). 13C-NMR (CDCl3): δ 173.7, 156.0, 137.3, 134.6, 129.7, 129.6, 125.7, 125.4, 115.7, 54.4, 50.4, 46.8, 44.0, 40.5, 27.3, 20.9.
Example 119 - N-Phenethyl-N-d-methylpiperidin-4-yl')-Nr-phenyl-carbamide (47 AKU- 30)
[0512] 4-(2-Phenylethyl)amino-l-methylpiperidine (110 mg, 0.5 mmol) was dissolved in 5 ml dichloromethane and placed in 50 ml flask. Phenylisocyanate (71 mg, 0.6 mmol) in 5 ml dichloromethane was added. After 16 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified twice by flash chromatography (0-10% methanol in dichloromethane) giving 131 mg (78%) 47AKU-30. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ =338.1 (UV/MS(%)=99/100). 1H-NMR (400 MHz, CDCl3): δ 7.36-6.93 (1OH, m); 6.24 (IH, s); 4.31 (IH, m); 3.50 (2H, t); 3.20 (2H, d); 2.89 (2H, t); 2.57 (2H, m); 2.50 (3H, s); 2.26 (2H, m); 1.79 (2H, m). 13C-NMR (CDCl3) δ 155.8, 139.2, 139.0, 129.4, 129.3, 128.9, 127.3, 123.2, 120.4, 54.9, 51.3, 45.5, 44.3, 37.6, 28.3.
Example 120 - N-(3-Phenylpropyl>N-π-methylpiperidin-4-ylVN'-benzyl-carbarnide (47AKU-3n
[0513] 4-(3-Phenylproρyl)amino-l-methylpiperidine(160 mg, 0.7 mmol) was dissolved in 5 ml dichloromethane and placed in 50 ml flask. Benzylisocyanate (107 mg, 0.8 mmol) in 5 ml dichloromethane was added. After 2 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified twice by flash chromatography (0-10% methanol in dichloromethane) giving 156 mg (61%) 47AKU-31. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.3. HPLC-MS (Method A): M+ =366.1 (UV/MS(%)=100/100). 1H-NMR (400 MHz, CDCl3): δ 7.34- 7.07 (10H5 m); 4.33 (3H, m); 4.14 (IH5 m); 3.04 (2H, m); 2.89 (2H5 d); 2.57 (2H5 1); 2.28 (3H, s); 2.06 (2H, m); 1.87 (2H, m); 1.75-1.62 (4H, m). 13C-NMR (CDCl3): δ 157.5, 141.0, 140.0, 129.0, 128.6, 128.3, 128.0, 127.6, 126.6, 55.6, 52.1, 46.3, 45.1, 41.6, 33.4, 32.2, 30.6.
Example 121 - N-r3-Phenylpropyl)-N-π-methylpiperidin-4-yl)-N'-phenyl-carbamide (47AKXJ-32)
[0514] 4-(3-Phenylpropyl)amino-l-methylpiperidine (160 mg, 0.7 mmol) was dissolved in 5 ml dichloromethane and placed in 50 ml flask. Phenylisocyanate (95 mg, 0.8 mmol) in 5 ml dichloromethane was added. After 20 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 106 mg (43%) 47AKU-32. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.3. HPLC-MS (Method A): M+ =352.1 (UV/MS(%)=100/100). 1H-NMR (400 MHz, CDCl3): δ 7.35-6.95 (1OH, m); 5.99 (IH, s); 4.18 (IH, m); 3.17 (2H, t); 2.91 (2H, d); 2.65 (2H, t); 2.28 (3H, s); 2.07 (2H, m); 1.97 (2H, m); 1.81-1.66 (4H5 m). 13C-NMR (CDCl3): δ 154.9, 141.0, 139.3, 129.2, 129.0, 129.0, 126.8, 123.1, 120.0, 55.6, 52.2, 46.2, 41.8, 33.4, 32.3, 30.6.
Example 122 - 2-(4-MethoxyphenylV2.2-ethylene-N-f4-methylbenzyl')-N-π- methylpiperidin-4- vDacetamide (47AKU-33')
[0515] l-(4-Methoxyphenyl)-l -cyclopropane carboxylic acid (230 mg, 1.2 mmol) was dissolved in 2 ml thionylchloride and placed in 50 ml flask. Mixture was heated to reflux for 2 hrs and then concentrated on Rotavapor (40° C). The acid chloride (250 mg, 1.2 mmol) in 5 ml dichloromethane was added to 47AKU-5 (220 mg, 1.0 mmol) in 5 ml dichloromethane. After 2 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified twice by flash chromatography (0-10% methanol in dichloromethane) giving 201 mg (51%) 47AKU-33. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.6. HPLC-MS (Method A): M+ =393.2 (UV/MS(%)=95/88).
105161 1H-NMR (400 MHz, CDCl3, rotamers): δ 7.22-6.70 (8H, m); 4.44 (2H, s); 4.26 (IH, m); 3.74 (3H, s); 3.12 and 2.89 (2H, 2m); 2.51 (IH, m); 2.32 (3H5 m); 2.26 (3H5 s); 2.08-1.52 (4H, m); 1.36 (2H, bs); 1.15-0.95 (3H, m). 13C-NMR (CDCl3) δ 172.9, 158.6, 136.6, 132.7, 129.2, 128.6, 127.9, 127.4, 114.4, 55.5, 55.1, 54.4, 45.2, 45.0, 29.8, 29.2, 21.2, 13.8.
Example. 123 - 2-(4-MethoxyphenvO-N-(l-phenylethy IVN-C 1-meth ylpiperidin-4- vDacetamide (47AKU-37)
4- Alpha-methylbenzylamino- 1 -methyl-piperidine ( 47 AKU-36")
[0517] DL-Phenylethylamine (606 mg, 5.0 mmol) was dissolved in 10 ml methanol and l-Methyl-4-piperidone (566 mg, 5.0 mmol) in 10 ml methanol was added. Mixture was stirred and Acetic acid (-0.75 ml) was added until pH~5. NaCNBH3 (628 g, 10 mmol) was slowly added. Gas evolution observed. After magnetic stirring for 20 hrs methanol was partly removed on Rotavapor (40° C). Ethylacetate, 2M NaOH and water were added until pH~10. Phases were separated and aq. phase was then re-extracted with ethylacetate and dichloromethane. Combined organic phases were dried over MgSO4. Concentration on Rotavapor (40° C.) yielded 838 mg crude 47AKU-36. TLC (10% methanol in dichloromethane): Rf =0.3. HPLC-MS (Method A): M+ =219.1 (UV/MS(%)=100/94).
2-(4-Methoxyphenyl")-N-alpha-methylbenzyl-N-π-methylpiperidin-4-yl)acetamid e (47AKU-37)
[0518] 47AKU-36 (218 mg, 1.0 mmol) was dissolved in 10 ml dichloromethane and placed in 50 ml flask. 4-Methoxyphenylacetyl chloride (185 mg, 1.2 mmol) in 10 ml dichloromethane was added. After 16 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 256 mg (70%) 47AKU-37. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.5. HPLC-MS (Method A): M+ =367.3 (UV/MS(%)=100/99).
105191 1H-NMR (400 MHz, CDCl3, rotamers): δ 7.34-7.06 (7H, m); 6.84 (2H, d); 5.10 (IH, m); 3.77 (3H, s); 3.67 (2H, m); 3.17 (IH, m); 3.03-2.75 (3H, m); 2.64 (3H, s); 2.38 (2H, m); 1.77-1.05 (6H, m). 13C-NMR (CDCl3): δ 172.0, 158.9, 139.9, 130.0, 129.0, 128.2, 127.1, 114.5, 55.5, 53.1, 51.4, 42.4, 41.3, 31.1, 29.5, 24.9, 18.1.
Example 124 - 2-f4-Methoxyphenyl)-N-r4-methylbenzyl)-N-(r8-methyl-8-aza- bicvclor3.2.11octen-3 -vH-acetamide f47AKU-39^ 4-Methylbenzylimino-tropane f 47AKU-38)
[0520] 4-Methylbenzylamine (1.21 g, 10 mmol) and Tropinone (1.39 g, 10 mmol) were placed in 100 ml flask and dissolved in 50 ml toluene. Mixture was heated to reflux for 3 hrs and water was removed using a Dean/Stark water-separator. Crude product was concentrated on Rotavapor (40° C.) giving 47AKU-38. TLC (10% methanol in dichloromethane): Rf =0.3. 1H-NMR (400 MHz, CDCl3, isomers): 7.20-7.09 (4H, m); 4.47 (IH, m); 3.81 (IH, s); 3.42 (IH, m); 3.31 (IH, m); 2.77-2.56 (2H, m); 2.47 and 2.41 (3H, 2s); 2.33 and 2.31 (3H, 2s); 2.27-1.97 (4H, m); 1.69-1.54 (2H, m).
2-("4-Methoxyphenvπ-N-(4-methylbenzylVN-(8-methyl-8-aza-bicvclof3.2.1]octen- 3-ylVacetamide f47AKU-39>l
[0521] 47AKU-38 (242 mg, 1.0 mmol) was dissolved in 5 ml dichloromethane and placed in 50 ml flask. 4-Methoxyphenylacetyl chloride (185 mg, 1.2 mmol) in 10 ml dichloromethane was added. After 16 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 69 mg (18%) 47AKU-39. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ =391.2 (UV/MS(%)=91/86).
|0S22J 1H-NMR (400 MHz, CDCl3, rotamers): δ 7.22-6.82 (8H, m); 5.41 (IH, bs); 4.71-4.52 (2H, m); 3.78 (3H, s); 3.68 (2H, m); 3.44-3.24 (2H, m); 2.72-2.36 (5H, m); 2.32 (3H, s); 2.25-2.00 (2H, m); 1.80-1.54 (2H, m). 13C-NMR (CDCl3): δ 170.8, 158.7, 137.4, 134.9, 130.1, 129.3, 128.9, 126.9, 114.2, 59.0, 58.0, 55.5, 49.5, 46.3, 39.7, 35.9, 33.8, 29.7, 21.3.
Example 125 - 2-Phenyl-2-ethyl-N-(4-methylbenzylVN-(l-methylpiperidin-4- yDacetamide f47AKTJ-40s)
[0523] 2-Phenylbutyric acid (197 mg, 1.2 mmol) was dissolved in 2 ml thionylchloride and placed in 50 ml flask. Mixture was heated to reflux for 2 hrs and then concentrated on Rotavapor (50° C). The acid chloride (1.2 mmol) in 5 ml dichloromethane was added to 47AKU-5 (158 mg, 0.72 mmol) in 5 ml dichloromethane. After 20 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 196 mg (74%) 47AKU-40. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.5. HPLC-MS (Method A): M+ =365.4 (UV/MS(%)=99/100). 1H-NMR (400 MHz5 CDCI3, rotamers): δ 7.32-6.98 (8H, m); 4.77 (IH5 bs); 4.50 (IH5 d); 4.29 (IH5 d); 3.43 and 3.21 (3H, 2m); 2.72 (2H, m); 2.62 (3H, s); 2.43 (IH5 m); 2.32 (3H5 s); 2.2 (3H5 m); 2.04 (2H5 m); 1.67 (3H, m); 0.92-0.72 (3H5 m). 13C-NMR (CDCl3): δ 174.7, 139.9, 137.3, 135.2, 129.7, 129.O5 127.8, 127.3, 125.8, 54.5, 51.6, 49.4, 46.0, 43.8, 28.9, 26.7, 26.3, 21.2, 12.7.
Example 126 - (47AKU-44~)N-(4-Methylbenzvn-N-f l-methylpiperidin-4-vn-N'-(4- methoxybenzyl Vcarbamide (47AKU-44)
[0524] 47AKU-5 (219 mg, 1.0 mmol) was dissolved in 5 ml dichloromethane and placed in 50 ml flask. 4-Methoxybenzylisocyanate (196 mg, 1.2 mmol) in 10 ml dichloromethane was added. After 16 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 192 mg (50%) 47AKU-44. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf=0.3. HPLC-MS (Method A): M+ =382.3 (UV/MS(%)= 100/94). 1H- NMR (400 MHz, CDCl3): δ 7.10 (4H5 m); 6.98 (2H5 m); 6.76 (2H5 m); 4.58 (IH5 1); 4.45 (IH5 m); 4.33 (2H5 s); 4.25 (2H5 d); 3.76 (3H5 s); 2.97 (2H5 m); 2.34 (3H, s); 2.32 (3H, s); 2.24 (2H5 m); 1.78 (4H, m). 13C-NMR (CDCl3): δ 158.9, 158.5, 137.3, 135.2, 131.8, 129.8, 128.8, 126.2, 114.1, 55.5, 55.4, 51.7, 45.8, 45.7, 44.5, 29.7, 21.2.
Example 127 - 2-(3,4-dimethoxyphenvI)-N-(4-meΦylbenzylVN-d-methylpiperidin-4- vDacetam ide T47AKU-45)
[0525] 3,4-Dimethoxyphenylbutyric acid (235 mg, 1.2 mmol) was dissolved in 2 ml thionylchloride and placed in 50 ml flask. Mixture was heated to reflux for 2 hrs and then concentrated on Rotavapor (50° C). The acid chloride (1.2 mmol) in 5 ml dichloromethane was added to 47AKU-5 (219 mg, 1.0 mmol) in 10 ml dichloromethane. After 16 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 129 mg (33%) 47AKU-45. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ =397.4 (UV/MS(%)=98/89). 1H-NMR (400 MHz, CDCl3, rotamers): δ 7.17-6.60 (7H5 m); 4.75 (IH5 m); 4.51 (2H5 s); 3.83 (3H5 s); 3.79 (3H5 s); 3.53 (2H5 s); 3.27 (2H5 d); 2.65 (2H, t); 2.58 (3H, s); 2.32 (3H, s); 2.24 (2H5 m); 1.72 (2H5 d). 13C-NMR (CDCl3): δ 172.8, 149.3, 148.3, 137.4, 135.0, 129.8, 127.4, 125.8, 121.05 112.2, 111.6, 56.2, 56.1 , 54.6, 49.6, 46.7, 44.0, 40.9, 27.0, 21.2.
Example 128 - 2-f3,4-Methylenedioxyphenyl')-N-r4-methylbenzyl')-N-(l-methylpiperidin- 4-yl)a cetamide f47AKU-46)
[0526] 3,4-Methylenedioxyphenylacetic acid (216 mg, 1.2 mmol) was dissolved in 2 ml thionylchloride and placed in 50 ml flask. Mixture was heated to reflux for 2 hrs and then concentrated on Rotavapor (50° C). The acid chloride (1.2 mmol) in 5 ml dichloromethane was added to 47AKU-5 (219 mg, 1.0 mmol) in 10 ml dichloromethane. After 2 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 188 mg (49%) product. Further purification by ion exchange chromatography (washout with 10% aq. NH4 OH (25%) in methanol) yielded 149 mg (39%) 47AKU-46. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ =381.2 (UV/MS(%)=96/95). 1H-NMR (400 MHz, CDCl3, rotamers): δ 7.17-7.02 (4H, m); 6.77-6.51 (3H5 m); 5.91 and 5.93 (2H, 2s); 4.70 (IH,- m); 4.52 and 4.49 (2H, 2s); 3.51 (2H,.s); 3.26 (2H, d); 2.49 (3H, s); 2.33 (3H5 s); 2.14-1.66 (6H, m) 13C-NMR (CDCl3): δ 172.5, 148.1, 146.8, 137.3, 135.1, 129.8, 128.6, 125.8, 121.9, 109.4, 108.5, 101.2, 54.8, 50.2, 46.7, 44.6, 41.1, 27.7, 21.2.
Example 129 - 2-f4-Methoxyphenyl)-N-r4-methylbenzyl)-N-(l-t-butylpiperidin-4-yl)- acetamid e f47AKU-49> l-t-Butyl-4-piperidone (47AKU-47)
[0527] 1 -Benzyl-4-piperidone (1.89 g, 10 mmol) was dissolved in 15 ml acetone. Methyliodide (0.90 ml, 15 mmol) was slowly added over 5 min. After 2 hrs magnetic stirring additional Methyliodide (1.8 ml, 30 mmol) was added. After 1 hr magnetic stirring 20 ml diethyl-ether was added. Crude product was collected by filtration and washed with acetone/diethyl ether. White crystals were dried under vacuum giving 806 mg quartenary salt. TLC (10% methanol in dichloromethane): Rf =0.7. Partly dissolved salt in 5 ml water was added to 50° C. hot mixture of t-Butylamine (120 mg, 1.6 mmol) and Potassiumcarbonate (32 mg, 0.22 mmol) in 3 ml ethanol. The resulting mixture was stirred and heated to reflux (~80° C.) for 1 hr. After cooling water (20 ml) and dichloromethane (20 ml) were added. Phases were separated and aq. phase was re- extracted with dichloromethane and ethylacetate. Combined organic phases were dried over MgSO4 and concentrated on Rotavapor (40° C.) giving 496 mg 47AKU-47. TLC (10% methanol in dichloromethane): Rf=0.3. 1H-NMR (400 MHz, CDCl3): δ 2.82 (4H31); 2.41 (4H5 t); 1.12 (9H, s). 13C-NMR (CDCl3): δ 210.2, 54.3, 46.4, 42.4, 26.6. Crude product contained ~25% (1H-NMR) starting material (l-Benzyl-4-piperidone).
4-(4-MethylbenzylaminoV 1 -t-butyl-piperidine (47 AKU-48)
[0528] 4-Methylbenzylamine (268 mg, 2.2 mmol) was dissolved in 5 ml methanol and placed in 50 ml flask. 47AKU-47 (305 mg, 2.0 mmol) in 5 ml methanol was added. Acetic acid (0.3 ml) was added until pH~5. NaCNBH3 (250 mg, 4.0 mmol) was slowly added. Gas evolution observed. After 4 hrs magnetic stirring dichloromethane, 2M NaOH and water were added until pH~10. Phases were separated and aq. phase was then re-extracted with dichloromethane and ethylacetate. Combined organic phases were dried over MgSO4. Concentration on Rotavapor (40° C.) yielded 556 mg crude 47AKU- 48. TLC (20% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ =261.2 (MS(%)=57).
2-(4-Methoxyphenyl')-N-(4-methylbenzyl)-N-π-t-butylpiperidin-4-yl)-acetamide (47AKU-49)
[0529] 47AKU-48 (556 mg, 2.1 mmol) was placed in 50 ml flask and 5 ml dichloromethane was added. 4-Methoxyphenylacetyl chloride (739 mg, 4.0 mmol) in 10 ml dichloromethane was added. After 4 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 124 mg (15%) product. Further purification by ion exchange chromatography (washout with 10% aq. NH4 OH (25%) in methanol) gave 91 mg (11%) 47AKU-49. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.5. HPLC-MS (Method A): M+ =409.4 (UV/MS(%)=100/90). 1H-NMR (400 MHz, CDCl3): δ 7.11 (4H, m); 7.03 (2H, d); 6.79 (2H5 d); 4.78 (IH, m); 4.56 (2H, s); 3.76 (3H, s); 3.53 (2H, s); 3.43 (2H, m); 2.63 (2H, m); 2.47 (2H, m); 2.31 (3H, s); 1.74 (2H, d); 1.36 (9H, s). 13C-NMR (CDCl3): δ 173.0, 158.8, 137.1, 135.3, 129.8, 129.7, 127.0, 125.8, 114.3, 55.6, 55.5, 49.8, 46.5, 46.4, 40.5, 26.7, 25.1, 21.2.
Example 130 - N-f4-Methylbenzyl')-N-('l-methylpiperidin-4-yl')-N'-phenylethyl-carbamide (58AKU-1)
105301 47AKU-5-2 (219 mg, 1.0 mmol) was dissolved in 5 ml dichloromethane and placed in 50 ml flask. Phenethylisocyanate (177 mg, 1.2 mmol) in 5 ml dichloromethane was added. After 6 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-15% methanol in dichloromethane) giving 134 mg (37%) 58AKU-1. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.5. HPLC-MS (Method A): M+ =366.3 (UV/MS(%)=99/96). 1H- NMR (400 MHz, CDCl3): δ 7.21-6.97 (9H, m); 4.33 (IH, m); 4.26 (IH, m); 4.21 (2H, s); 3.39 (2H, q); 2.85 (2H, m); 2.67 (2H5 1); 2.31 (3H, s); 2.24 (3H, s); 2.06 (2H, m); 1.73- 1.57 (4H, m). 13C-NMR (CDCl3): δ 158.7, 139.5, 137.0, 135.4, 129.7, 128.8, 128.6, 126.3, 126.1, 55.6, 52.2, 46.2, 45.8, 42.2, 36.4, 30.2, 21.2.
Example 131 - N-Phenylethyl-N-fl-methylpiperidin^-vD-N'-phenethyl-carbamide (58AKU-2)
[0531] 4-(2-Phenylethyl)amino-l-methylpiperidine (131 mg, 0.6 mmol) was dissolved in 5 ml dichloromethane and placed in 50 ml flask. Phenethylisocyanate (103 mg, 0.7 mmol) in 5 ml dichloromethane was added. After 4 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (45° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 198 mg (90%) 58 AKU-I. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.3. HPLC-MS (Method A): M+ =366.3 (UV/MS(%)=100/100). 1H-NMR (400 MHz, CDCl3): δ 7.33-7.16 (8H, m); 7.01 (2H5 m); 4.23 (IH, t); 4.04 (IH, m); 3.47 (2H, q); 3.17 (2H, t); 2.89 (2H5 m); 2.78 (2H, t); 2.66 (2H, t); 2.28 (3H, s); 2.05 (2H, m); 1.79-1.59 (4H, m). 13C-NMR (CDCl3): δ 157.8, 139.6, 139.0, 129.0, 128.9, 128.8, 126.8, 126.7, 55.7, 52.5, 46.2, 44.6, 42.0, 37.3, 36.4, 30.5. Example 132 N-f 4-MethylbenzylVN-f 1 -t-butylpiperidin-4-yl>N'-f 4- methoxybenzvBcarbamid e (58AKU-3)
[0532] 47AKU-5-2 (404 mg, 1.6 mmol) was dissolved in 5 ml dichloromethane and placed in 50 ml flask. 4-Methoxybenzylisocyanate (326 mg, 2.0 mmol) in 5 ml dichloromethane was added. After 20 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (45° C). Crude product was purified three times by flash chromatography (0-20% methanol in dichloromethane and 0-30% methanol in ethylacetate) giving 155 mg (23%) 58AKU-3. HCl-salt was prepared from 2M HCl/diethylether in dichlorom ethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.3. HPLC-MS (Method A): M+ =424.2 (UV/MS(%)=92/83). 1H-NMR (400 MHz, CDCl3): δ 7.10 (4H, m); 6.99 (2H5 m); 6.76 (2H, m); 4.53 (IH, m); 4.35 (3H, s); 4.26 (2H5 d); 3.77 (3H5 s); 3.09 (2H, m); 2.32 (3H3 s); 2.22 (2H, m); 1.81-1.54 (4H5 m); 1.06 (9H, s). 13C-NMR (CDCl3): δ 158.9, 158.6, 137.1. 135.6 131.9, 129.7, 128.8, 126.2, 114.0, 62.6, 55.5, 53.0, 45.9, 45.7, 44.5, 31.0, 26.3, 21.2.
Example 133 - 2-(4-EthoxyphenylVN-('4-methylben2ylVN-('l-methylpiperidin-4- vDacetamide f58AKU-4)
[0533] 4-Ethoxyphenylacetic acid (270 mg, 1.5 mmol) was dissolved in 2 ml thionylchloride and placed in 50 ml flask. Mixture was heated to reflux for 2 hrs and then concentrated on Rotavapor (45° C). The acid chloride (1.5 mmol) in 5 ml dichloromethane was added to 47AKU-5-2 (262 mg, 1.2 mmol) in 5 ml dichloromethane. After 20 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 272 mg (60%) 58AKU-4. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.4. HPLC-MS (Method A): M+ =381.2 (UV/MS(%)=98/91). 1H-NMR (400 MHz, CDCl3): δ 7.17-6.99 (6H, m); 6.82-6.76 (2H, m); 4.73 (IH, m); 4.48 (2H, s); 3.98 (2H, q); 3.52 (2H, s); 3.22 (2H, d); 2.61 (2H, t); 2.54 (3H, s); 2.32 (3H5 s); 2.14 (2H, s); 1.71 (2H, d): 1.38 (3H, t). 13C-NMR (CDCl3): δ 172.9, 158.2, 137.3, 135.0, 129.9, 129.8, 126.8, 125.8, 114.9, 63.7, 54.6, 49.8, 46.7, 44.1, 40.6, 27.2, 21.2, 15.0. Example 134 - 2-f4-Butoxyphenyl)-N-f4-methylbenzylVN-d-methylpiperidin-4- vnacetamide (58AKU-5)
[0534J 4-Butoxyphenylacetic acid (317 mg, 1.5 mmol) was dissolved in 2 ml thionylchloride and placed in 50 ml flask. Mixture was heated to reflux for 2 hrs and then concentrated on Rotavapor (45° C). The acid chloride (1.5 mmol) in 5 ml dichloromethane was added to 47AKU-5-2 (262 mg, 1.2 mmol) in 5 ml dichloromethane. After 20 hrs magnetic stirring the reaction mixture was concentrated on Rotavapor (40° C). Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) giving 230 mg (47%) 58AKU-5. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.5. HPLC-MS (Method A): M+ =409.2 (UV/MS(%)=98/93). 1H-NMR (400 MHz, CDCl3): δ 7.15-6.96 (6H, m); 6.78 (2H5 m); 4.74 (IH, m); 4.48 (2H, s); 3.91 (2H, t); 3.52 (2H, s); 3.27 (2H5 d); 2.72 (2H, t); 2.58 (3H, s); 2.32 (3H5 s); 2.23 (2H5 m); 1.72 (4H5 d); 1.45 (2H, m); 0.95 (3H, t). 13C-NMR (CDCl3): δ 173.0, 158.4, 137.3, 135.0, 129.8, 126.6, 125.8, 115.0, 67.9, 54.4, 49.5, 46.7, 43.8, 40.6, 31.5, 26.8, 21.2, 19.4, 14.0.
Example 135 - 2-(4-i-Propoxyphenyl')-N-('4-methylbenzyl)-N-ri-methylpiperidin-4- vnacetamid e (58AKU-6)
[0535] 47AKU-29-2 (245 mg, 0.7 mmol) was dissolved in 10 ml dimethylformamide and placed in 50 ml flask. KOH (196 mg, 3.5 mmol) and Isopropylbromide (200μl, 2.1 mmol) were added. Mixture was heated to 50° C. and stirred for 24 hrs. After cooling water and ethylacetate were added. Phases were separated and aq. phase was then re-extracted with dichloromethane. Combined organic phases were washed with brine, dried over MgSO4 and concentrated on Rotavapor (40° C.) giving 188 mg. Crude product was purified by flash chromatography (0-10% methanol in dichloromethane) yielding 136 mg (49%) 58AKU-6. HCl-salt was prepared from 2M HCl/diethylether in dichloromethane/heptane. TLC (10% methanol in dichloromethane): Rf =0.3. HPLC-MS (Method B): M+ =395 (UV/MS(%)=95/91). 1H-NMR (400 MHz, CDCl3, rotamers): δ 7.23-7.01 (6H, m); 6.79 (2H, m); 4.60 (IH, m); 4.51 (IH, m); 4.44 (IH, s); 3.77 (IH, s); 3.52 (IH, s); 2.83 (2H, m); 2.76 (2H, m); 2.28 and 2.34 (3H, 2s); 2.19 and 2.22 (3H5 2s); 2.05 (IH, m); 1.86-1.55 (4H, m); 1.32 (6H, d). 13C-NMR (CDCl3): δ 172.6, 157.0, 137.1, 135.6, 129.8, 129.7, 125.8, 116.2, 70.1, 55.3, 51.6, 46.6, 46.1, 40.8, 29.6, 22.3, 21.2.
Pharmalogical Data
Example 136 - Receptor Selection and Amplification (R-SAT) Assays.
[0536] The functional receptor assay, Receptor Selection and Amplification Technology (R-SAT), was used (with minor modifications from that previously described U.S. Pat. No. 5,707,798) to screen compounds for efficacy at the 5-HT2A receptor. The RSAT assay was conducted as described herein. The results obtained for several compounds of the invention are presented in Table 3, below.
Table 3. Efficiency And pIC50 Of Compounds At The 5-HT2A Receptor Compared To Ritanserin.
Figure imgf000154_0001
Figure imgf000155_0001
Example 137 - Selectivity Profile for 2-(4-MethoxyphenylVN-f4-methylbenzvn-N-ri- methylpiperidin-4-yDacetamide hydrochloride
[0537] The R-SAT assay (described above in example 136) was used to investigate the selectivity of 2-(4-Methoχyρhenyl)-N-(4-methylbenzyl)-N-(l- methylpiperidin-4-yl)acetamide hydrochloride. The results from a broad profiling of this compound at a variety of receptors are reported in Table 4 below. NR means No Response, i.e. the compound investigated showed no effect at the receptor studied.
Table 4. Selectivity of 2-(4-methoxyphenyl)-n-(4-methylbenzyl)-n-(l -methylpiperidin-4- yl) acetamide.
Figure imgf000156_0001
Figure imgf000157_0001
HPLC/LCMS Method for Examples 138 to 252
[0538] The analysis was performed on a combined prep/analytical
Waters/Micromass system consisting of a ZMD single quadropole mass spectrometer equipped with electrospray ionization interface. The HPLC system consisted of a Waters 600 gradient pump with on-line degassing, a 2700 sample manager and a 996 PDA detector. , Separation was performed on an X-Terra MS Cl 8, 5 μm 4.6x50mm column. Buffer A: 1OmM ammoniumacetate in water, buffer B: 1OmM ammoniuacetate in acetonitrile/water 95/5. A gradient was run from 30%B to 100%B in 7 min, hold at 100%B for 1 min and re-equilibrated for 5.5 min. The system was operated at 1 ml/min.
Preparation of hydrochloride salts.
[0539] Typically, the tertiary amines were dissolved in dichloromethane, treated with an excess of IM HCl in diethylether and precipitated from n-heptane. The solvents were removed in vacuo and after drying, the hydrochloride salts were obtained as colorless solids.
Preparation of oxalate or tartrate salts.
[0540] Typically, the tertiary amines were dissolved in methanol, treated with 1 eq. of the appropriate acid, the solvent removed and the salt redissolved in dichloromethane and precipitated from «-heptane. The solvents were removed in vacuo affording the salts as colorless solids.
Preparation of phenylacetyl chloride derivatives
[0541] The phenylacetic acid derivative (15 mmol) was dissolved in dichloromethane (100 mL), and oxalylchloride (45 mmol) was added slowly. The reaction mixture was stirred for 4 hours and then evaporated to dryness. The product was obtained as a colorless oil and used immediately after preparation in the acylation step.
Example 138 - 4-Isobutoxyphenylacetic acid (128NLS281
[0542] Methyl 4-hydroxyphenyl acetate (14.6 g, 0.0885 mol) was dissolved in DMF (200 mL), potassium carbonate (31.0 g, 0.224 mmol) added and the mixture was stirred for 1 h at rt. 1 -Bromo-2-methylpropane (19.2 mL, 0.177 mol) was added and the mixture was heated at 8O0C for 3 days under vigorous stirring. The mixture was cooled to rt. filtered, the solvent removed and the residue partitioned between 1.5M NaOH and ethyl acetate. The organic layer was evaporated, the residue dissolved in methanol (100 mL) and water (100 mL), KOH (10 g, 0.178 mol) added and the mixture stirred overnight at rt. The methanol was removed by evaporation, the mixture extracted with dichloromethane. The organic layer was discarded, the aqueous layer acidified with 4M HCl to pH2-3 and extracted twice with dichloromethane. The combined organic layers were dried over Na2SO^ filtered and evaporated to give the title compound (16.9 g, 92%) as a colorless solid.
Example 139 - 4-Propoxyphenylacetic acid (98AF77-66')
[0543] Prepared as described for 128NLS28 using propylbromide as the alkylating agent. Example 140 - 4-Isopropoχvphenylacetic acid (130AF24-163')
[0544] Prepared as described for 128NLS28 using isopropylbromide as the alkylating agent.
Example 141 - N-π-r2-π.3-Dioxolan-2-yl')ethvnpiperidin-4-vπ-N-r4-fluorobenzylVN'- (4-isobutoxybenzvDcarbamide. hydrochloride C80MBT86-2C)
[0545] 4-Piperidone hydrochloride monohydrate (4.0 g, 26.0 mmol) was dissolved in dichloromethane (130 mL). After addition of triethylamine (8.66 g, 85.8 mmol), the mixture was stirred for 10 min and then cooled to 00C. Trifluoroacetic anhydride (12.0 g, 57.2 mmol) was added dropwise under stirring. After 2 h at room temperature, the reaction was stopped by addition of water (100 mL), and the aqueous phase was extracted with dichloromethane (2x100 mL). The combined organic phases were dried over Na2SCM, filtered and concentrated to give l-(trifluoroacetyl)-4-piperidone (5.07 g, 100%).
[0546] 4-Fluorobenzylamine (3.14 g, 25.9 mmol) was dissolved in methanol (150 mL). l-(trifiuoroacetyl)-4-piperidone (5.07 g, 25.9 mmol) was added, and the pH was adjusted to ~5 with acetic acid. The reaction mixture was stirred for 5 min and NaBH3CN (2.46 g, 38.9 mmol) was added slowly under stirring. After 20 h at room temperature the reaction was concentrated. 2 M NaOH (100 mL) was added and the aqueous phase was extracted with dichloromethane (2x100 mL). The combined organic phases were dried over Na2SO4, filtered and concentrated to give N-(4-fluorobenzy I)-I- (trifluoroacetyl)piperidin-4-amine (5OELH85, 2.91 g, 37%).
[0547] 4-isobutoxyphenylacetic acid (7.6 g, 36.5 mmol) was dissolved in THF (50 mL). Proton Sponge™ (8.2 g, 38 mmol) was added, and the mixture was stirred for 15 min. Diphenylphosphoryl azide (10.6 g,' 38 mmol) was added dropwise and the mixture was heated to reflux for 4 h. The mixture was cooled to room temperature and placed in the freezer at -18 0C for 20 h. The resulting white precipitate was vigorously stirred with diethyl ether (250 mL) for 15 min and filtered. The filtrate was evaporated to give crude 4-isobutoxybenzyl isocyanate (1.97 g, 9.6 mmol), which was dissolved in dichloromethane (50 mL) and added to a solution of 50ELH85 (2.91 g, 9.6 mmol) in dichloromethane (50 mL). The reaction mixture was stirred for 20 h and concentrated. The crude product was purified by flash chromatography (0-5% methanol in dichloromethane) to give N-(4-fluorobenzyl)-N-[l-(trifluoroacetyl)piperidin-4-yl]-N'-(4- isobutyloxybenzyl)carbamide (76ELH17, 3.90 g, 91%).
[0548] The compound 76ELH17 (3.90 g, 8.7 mmol) was dissolved in methanol (12 ml) and added to a 2 M solution of potassium carbonate in methanol (100 mL) under stirring. After 4 h the methanol was evaporated, and the aqueous phase was extracted with dichloromethane (2x100 mL). The combined organic phases were dried over Na2SO4, filtered and concentrated to give a semi-pure solid (2.95 g), which was purified by flash chromatography (10% methanol in dichloromethane with 1% triethylamine) to give N-(4-fluorobenzyl)-N-(piperidin-4-yl)-N'-(4- isobutyloxybenzyl)carbamide (76ELH18, 1.40 g, 39%) as a colorless solid. LCMS m/z 414 [M+H]+. 1H-NMR (CDCl3): δ 7.21-6.75 (m, 8H)5 4.47-4.42 (m, IH), 4.39 (t, J=5 Hz, IH)3 4.35 (s, 2H), 4.27 (d, J=5 Hz, 2H), 3.68 (d, J=6 Hz, 2H), 3.13-3.06 (m, 2H), 2.74- 2.66 (m, 2H), 2.11-1.99 (m, IH), 1.78-1.71 (m, 3H), 1.58-1.46 (m, 2H), .1.00 (d, J=6 Hz, 6H).
[0549] The compound 76ELH18 (200 mg, 0.484 mmol) was dissolved in acetonitrile (20 mL). Potassium carbonate (74 mg, 0.553 mmol) and sodium iodide (80 mg, 0.553 mmol) was added followed by 2-(2-bromoethyl)-l,3-dioxolane (100 mg, 0.553 mmol). The reaction mixture was heated to reflux for 20 h. The mixture was concentrated, water (50 mL) was added, and the aqueous phase was extracted with dichloromethane (2x50 mL). The combined organic phases were dried over Na2SO4, filtered and evaporated. The resulting oil was purified twice by flash chromatography (5% methanol in dichloromethane) to give a colorless oil (50 mg, 20%). R/= 0.70 (MeOH/CH2Cl2 1:9). LCMS m/z 514 [M+H]+. 1H-NMR (CDCl3): δ 7.21-6.75 (m, 8H), 4.94 (t, j=4.5 Hz, IH), 4.73-4.62 (m, IH), 4.58 (t, J=5.5 Hz, IH)5 4.41 (s, 2H), 4.26 (d, J=5.5 Hz, 2H), 4.00-3.80 (m, 4H), 3.68 (d, J=6.0 Hz, 2H), 3.43-3.35 (m, 2H), 2.94-2.87 (m, 2H), 2.68-2.57 (m, 2H), 2.45-2.32 (m, 2H), 2.20-2.13 (m, 2H), 2.10-2.00 (m, IH), 1.88-1.81 (m, 2H), 1.00 (d, J=6.0 Hz, 6H). HPLC tκ = 8.1 min.
[0550] The collected compound was converted into its hydrochloride salt, which was obtained as a colorless solid (80MBT86-2C).
Example 142 - N-π-r2-(1.3-Dioxan-2-yl')ethyl1piperidin-4-yl>-N-r4-fluorobenzvn-2-r4- (2-hydroxy-2-methylpropoxy')phenyl]acetarnide, tartrate ("106MBT54-D) [0551] Methyl (4-hydroxyphenyl)acetate (500 mg, 3.0 mmol) was dissolved in DMF (3 mL). K2CO3 (829 mg, 6.0 mmol) was added followed by isobutylene oxide (800 μL, 9.0 mmol). The mixture was heated to 150 0C by microwave irradiation for 30 min and concentrated . The residue was dissolved in a 1:1 mixture of methanol and water (20 mL). NaOH (1 g) was added and the mixture was stirred for 30 min. Methanol was removed by rotary evaporation. The aqueous phase was acidified by 4 M HCl and extracted with dichloromethane (2x50 mL). The combined organic phases were extracted with 2 M NaOH (2x50 mL). The combined aqueous phases were subsequently acidified by 4 M HCl and extracted with dichloromethane (2x50 mL). The combined organic phases were dried over Na2SO4, filtered and evaporated to afford [4-(2-hydroxy-2- methylpropoxy)phenyl]acetic acid (106MBT52-D, 470 mg, 70%) as a colorless solid. 1H- NMR (CDCl3): δ 7.19 (m, 2H)3 6.88 (m, 2H), 3.78 (s, 2H)5 3.57 (s, 2H), 1.34 (s. 6H).
[0552] The acid 106MBT52-D (150 mg, 0.67 mmol) was dissolved in dichloromethane (10 mL). iV-{l-[2-(l,3-dioxan-2-yl)ethyl]piperidin-4-yl}-N-(4- fluorobenzyl)amine (118AF52-95, 180 mg, 0.56 mmol) was added followed by triethylamine (235 μL, 0.84 mmol). Bromo-tris-pyrrolidino-phosphonium hexafluorophosphate (PyBroP, 392 mg, 0.84 mmol) was added, and the mixture was stirred at room temperature for 2 h. The mixture was concentrated and passed onto a prewashed (methanol) ion exchange column (0.88 mmol/g, Ig). The column was washed with methanol (8x4 mL) and the remaining product was eluted off the column with 10% NH4OH in methanol (2x4 mL) and evaporated. The resulting oil was dissolved in dichloromethane (20 mL) and washed with saturated aqueous NaHCO3 (5x20 mL). The organic phase was dried over Na2SO4, filtered and evaporated. The resulting oil was purified by flash chromatography (0-5% methanol in dichloromethane) to give a colorless oil (110 mg, 31%). R/ = 0.64 (MeOHZCH2Cl2 1:9). LCMS w/z 529 [M+H]+. 1H-NMR (CDCl3, rotamers 0.4:0.6): δ 7.25-6.82 (m, 8H)5 4.64-4,48 (m, 2.4H), 4.44 (s, 1.2H), 4.10- 4.03 (m, 2H), 3.79-3.67 (m, 5.2H), 3.50 (s, 1.2H), 2.90-2.81 (m, 2H), 2.42-3.95 (m, 2H), 2.12-1.98 (m, 2.2H), 1.87-1.79 (m, 0.8H), 1.76-1.48 (m, 5.2H), 1.36-1.27 (m, 7.8H). HPLC /R = 6.1 min.
[0553] The collected compound was converted into its tartrate salt, which was obtained as a colorless solid (106MBT54-D). Example 143 - N-r4-FluorobenvzlVN-('piperidin-4-yl')-2-(/4-isobutoxyphenvπacetamide α03NLS56V
[0554] To a solution of the amine 118AF93-51 (10.37g, 30.3 mmol) and triethylamine (9.36 mL, 60.6 mmol) in dichloromethane (200 raL) a solution of 4- isobutoxyphenylacetyl chloride 128NLS28 (8.93 g, 39.4 mmol) in dichloromethane (100 mL) is added dropwise at O0C. The solution is stirred at rt for 3h, then water is added and the mixture washed with sat. aq. NaHCO3. The organic layer was washed with 5% HCl, water and brine, dried over sodium sulfate, filtered and evaporated in vacuo. The residue was purified by silica gel column chromatography eluting with a stepwise gradient of 0- 50% ethyl acetate in n-heptane, affording N-(4-fluorobenyzl)-iV-[l- (benzyloxycarbonyl)piperidin-4-yl]-2-(4-isobutoxyphenyl)acetamide as a colorless oil.
[0555] This compound was dissolved in abs. ethanol (200 mL) and hydrogenated overnight at rt using Pd/C (10%, 1 g) as a catalyst. The mixture was filtered over Celite, the solvent removed and the residue dried in vacuo to give a colorless oil (7.02 g, 58% over both steps). This compound was used without further purification. LCMS m/z 399 [MH-H]+. HPLC /R = 8.8 min.
Example 144 - iV^ll-fS-π.S-Dimethylpiperidin-l-vDpropylipiperidin^-vU-N-^- fluorobenzylV2-(4-isobutoxyphenyl)acetamide. dihvdrochloride (103NLS45-B').
[0556] To 3,5-dimethylpiperidine (43 μL, 0.33 mmol) in DMF (1 mL) was added potassium carbonate (132 mg, 1.0 mmol), followed by l-chloro-3-iodopropane (32 μmol, 0.30 mmol) and the mixture stirred at 500C for 2h. After cooling to rt, a solution of 103NLS56 (100 mg, 0.25 mmol) in DMF (0.5 mL) was added, followed by sodium iodide (45 mg, 0.30 mmol). The mixture was shaken for 20 h at 6O0C, filtered, evaporated to dryness and purified by silica gel column chromatography, eluting with a stepwise gradient of 0-10% methanol in dichloromethane. The residue was further purified by passage over a reversed phase Ci % SPE cartridge, giving the desired compound (35 mg, 25%), which was converted into its dihydrochloride salt.
[0557] R/= 0.61 (MeOHZCH2Cl2 1 :9). LCMS m/z 552 [M+H]+. HPLC /R = 8.7 min.
Example 145 - l-r3-(4-U4-Fluoroben2ylVf2-(4-isobutoxyphenyl')acetyl1aminolpiperidin- l-yPpropyπpiperidine-4-carboxyIic acid methyl ester, dihvdrochloride π03NLS45-E\ [0558] Prepared following the same method as described for 103NLS45-B, using piperidine-4-carboxylic acid methyl ester (44 μL, 0.33 mmol). Yield: 7 mg, 5%. [0559] LCMS m/z 582 [M+H]+. HPLC tR = 7.8 min.
Example 146 - N-(4-FluorobenzylV2-(4-isobutoxyphenyl')-N-π-r2-(l-methylpyrrolidin- 2-yl)ethvIlpiperidin-4-yl)acetamide. dioxalate C103NLS63-G).
[0560] To a solution of the amine 103NLS56 (15 mg, 0.038 mmol) in DMF (0.3 mL) was added a solution of 2-(2-chloroethyl)-l-methylpyrrolidine hydrochloride (8.4 mg, 0.045 mmol) in DMF (0.1 mL), followed by caesium carbonate (50 mg, 0.15 mmol) and sodium iodide (6.8 mg, 0.045 mmol). The mixture was stirred overnight at 6O0C, partitioned between dichloromethane and sat. aq. NaHCO3 solution. The organic layer was dried over sodium sulfate, filtered and evaporated. The residue was purified by preparative reversed phase (CJS) HPLC and the obtained compound (10.5 mg, 54%) converted into its dioxalate salt.
[0561] LCMS m/z 510 [M+H]+. HPLC fR = 8.1 min.
Example 147 - N-(l-|'3-r2,6-Dimethylmorpholin-4-yl')propyl'|piperidin-4-yl>-N-r4- fluorobenzviy2-f4-isobutoxyphenyl)acetamide. dioxalate (103NLS69-A\
[0562] To a solution of 2,6-dimethylmorpholine (6.1 μL, 49 μmol) in DMF (0.3 mL) l-chloro-3-iodopropane (4.9 μL, 45 μmol) in DMF (0.05 mL) was added, followed by caesium carbonate (50 mg, 0.15 mmol). The mixture was shaken at 5O0C for 3 h. After cooling to rt, the piperidine derivative 103NLS56 (15 mg, 38 μmol) in DMF (0.1 mL) and sodium iodide (6.8 mg, 45 μmol) were added and stirring maintained overnight at 600C. The mixture was partitioned between dichloromethane and sat. aq. NaHCC>3 solution. The organic layer was dried over sodium sulfate, filtered and evaporated. The residue was purified by preparative reversed phase (C18) HPLC and the obtained compound (6.3 mg, 30%) converted into its dioxolate salt.
[0563] LCMS m/z 554 [M+H]+. HPLC tR = 8.7 min.
Example 148 - N-(4-Fluorobenzyl)-N-{l-f3-r3-hydroxypiperidin-l-yl>)propynpiperidin-4- yl)-2-f4-isobutoxyphenyl)acetamide. dioxalate (T03NLS69-B). [0S64] Prepared following the same method as described for 103NLS69-A, using 3-hydroxypiperidine hydrochloride (6.8 mg, 49 μmol). Yield: 7.9 mg, 30%. LCMS m/z 540 [M+H]+. HPLC /R = 8.1 min.
Example 149 - N-(4-Fluoroben2yl)-2-f4-isobutoxyphenvO-N-π-P-(2-methylpiperidin-l- yl)propyl]piperidin-4-yl}acetamide, dioxalate (103NLSOg-C).
[0565] Prepared following the same method as described for 103NLS69-A, using 2-methylpiperidine (5.8 μL, 49 μmol). Yield: 5.2 mg, 26%. LCMS m/z 538 [M+H]+. HPLC fR = 8.7 min.
Example 150 - N-(4-Fluorobenzyl)-2-(4-isobutoxyphenyO-N-fl-f3-pyrrolidin-l-yl- propyl)piperidin-4-yl'jacetamide, dioxalate (103NLS69-D
[0566] Prepared following the same method as described for 103NLS69-A, using pyrrolidine (5.0 μL, 49 μmol). Yield: 4.6 mg, 24%. LCMS m/z 510 [M+H]+. HPLC tR = 8.4 min.
Example 151 - N-π-[3-("2,5-Dimethylpyrrolidin-l-yl)propyl1piperidin-4-yl>-N-(4- fluorobenzvP-2-(4-isobutoxyphenyl)acetamide,, dioxalate (103NLS69-E).
[0567] Prepared following the same method as described for 103NLS69-A, using 2,5-dimethylpyrrolidine (6.0 μL, 49 μmol). Yield: 3.4 mg, 17%. LCMS m/z 538 [M+H]+. HPLC /R = 8.7 min.
Example 152 - N-(4-Fluorobenzyl)-N-(l-f3-(3-hvdroxymethylpiperidin-l- yl)propyl1piperidin-4-yl)-2-(4-isobutoxyphenyl)acetamide< dioxalate Cl 03NLS69-F).
[0568] Prepared following the same method as described for 103NLS69-A, using 3-hydroxyrnethylpiperidine (5.5 μL, 49 μmol). Yield: 5.5 mg, 26%. LCMS m/z 554 [M-I-H]+. HPLC tκ = 8.0 min.
Example 153 - f4S)-3-(3-chloropropyl)-4-isopropyloxazolidinon-2-one (103NLS94).
[0569] Sodium hydride (60% suspension in oil, 288 mg, 7.2 mmol) was added to a solution of (5)-4-isopropyl-2-oxazolidinone (775 mg, 6.0 mmol) in dry tetrahydrofuran (50 mL) under argon atmosphere. The suspension was stirred for 15 min at rt, then l-bromo-3-chloropropane (1.18 mL, 12.0 mmol) was added dropwise over 30 min. The mixture was refluxed overnight, filtered and the filtrate evaporated in vacuo. Purification of the residue by silica gel column chromatography, eluting with a stepwise gradient of 0-4% methanol in dichloromethane afforded (45)-3-(3-chloropropyl)-4- isopropyloxazolidinon-2-one (824 mg, 67 %) as a colorless oil.
Example 154 - N-r4-Fluoroben2ylV2-r4-isobutoxyphenylVN-{l-f3-r4-('S)-isopropyl-2- oxo-oxazolidin-3-yl)propyπpiperidin-4-yUacetamide, oxalate d lTNLSOl).
[0570] To a solution of 103NLS56 (207 mg, 0.52 mmol) potassium carbonate (215 mg, 1.56 mmol) was added, followed by the alkylating agent 103NLS94 (127 mg, 0.62 mmol) and sodium iodide (93 mg, 0.62 mmol). The mixture was stirred at 65°C overnight, the solvent removed and the residue partitioned between ethyl acetate and water. The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by silica gel column chromatography, eluting with a stepwise gradient of 0-4% methanol in dichloromethane. Further purification of the compound was performed by passage over an acidic ion exchange SPE cartridge, affording the desired compound (209 mg, 71%) as a colorless oil, which was converted into its oxalate salt.
[0571] R/ = 0.35 (MeOH/CH2Cl2 5:95).. LCMS m/z 568 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.21-6.80 (m, 8H, Ar-H)5 4.60-4.53 (m, 0.6H, pip-H), 4.49 and 4.43 (2s, 2H, benzyl-H), 4.19-4.14 (m, IH, oxa-CH2), 4.06-4.01 (m, IH, oxa-CH2), 3.77-3.67 (m, 4.2H5 pip-H, oxa-NCH, CH2OiBu, benzyl-H), 3.53-3.46 (m, 2.2H, benzyl-H, OCONCH2), 2.98-2.85 (m, 3H, pip-H, OCONCH2), 2.39-2.25 (m, 2H, NCH2), 2.10-2.00 (m, 3.2H, CH(CHs)2, pip-H, CHOiBu), 1.85-1.50 (m, 6H, pip-H, NCH2CH2), 1.29 (m, 0.8H, pip-H), 1.01-0.99 (m, 6H, CH3OiBu), 0.89-0.83 (m, 6H, CH(CHs)2). ΗPLC tR = 8.9 min.
Example 155 - N-r2-r4-Fluoroρhenyl)ethyl1-2-r4-isobutoxyphenvn-N-π-r3-r4-CS)- isopropyl-2-oxo-oxazolidin-3-vπpropyllpiperidin-4-vπacetamide, oxalate (1 17NLS03-
[0572] Prepared following the same method as described for 117NLS01 using -V-[2-(4-fluorophenyl)ethyl]-2-(4-isobutoxyphenyl)-Λr-(piperidin-4-yl)acetamide (111 mg, 0.27 mmol, prepared by the procedure described for 103NLS56). Yield: 90 mg, 57%. [0573] R/ = 0.30 (MeOH/CH2Cl2 5:95). LCMS m/z 582 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.18-6.80 (m, 8H, Ar-H)5 4.40-4.35 (m, 0.4H, pip-H), 4.20- 4.15 (m, IH, oxa-CH2), 4.05-4.01 (m, IH, oxa-CH2), 3.75-3.46 (m, 6.6H5 pip-H, oxa- NCH, CH2OiBu, benzyl-H, OCONCH2), 3.36 (m, 2H5 ArCH2CH2N), 3.02-2.84 (m, 3H5 pip-H, OCONCH2), 2.81-2.75 (m5 2H, ArCH2), 2.37-2.25 (m, 2H, NCH2), 2.09-1.98 (m5 2.8H, CH(CH3)2, pip-H, CHOIBU), 1.85-1.62 (m, 6H, pip-H, NCH2CH2), 1.31 (m, 1.2Η, pip-H), 1.00-0.97 (m, 6H, CH3OiBu), 0.89-0.84 (m, 6H, CH(CHa)2). ΗPLC rR = 9.1 min.
Example 156 - N-[2-r4-Fluorophenvπethyl]-N-| l-[3-('4-(SVisopropyl-2-oxo-oxazolidin- 3-yl)propyl1piperidin-4-vU-2-(4-propoxyphenyl)acetamide, oxalate (117NLS03-B).
[0574] Prepared following the same method as described for 117NLS01 using iV-[2-(4-fluorophenyl)ethyl]-iV-(piperidin-4-yl)-2-(4-propoxyphenyl)acetamide (108 mg, 0.27 mmol, prepared by the procedure described for 103NLS56). Yield: 76 mg, 50%.
[0575] R/ = 0.33 (MeOΗ/CΗ2Cl2 5:95). LCMS m/z 568 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.17-6.81 (m, 8H, Ar-H), 4.40-4.35 (m, 0.4H, pip-H), 4.20- 4.15 (m, IH5 oxa-CH2), 4.05-4.01 (m, IH, oxa-CH2), 3.90-3.85 (m, 2H, OCH20Pr), 3.72- 3.48 (m, 4.6H, pip-H, oxa-NCH, benzyl-H, OCONCH2), 3.36-3.30 (m, 2H, ArCH2CH2N), 2.99-2.86 (m, 3H5 pip-H, OCONCH2), 2.80-2.74 (m, 2H5 ArCH2), 2.38-2.26 (m, 2H5 NCH2), 2.1 1-2.03 (m, 1.8H, CH(CH3)2, pip-H), 1.87-1.64 (m, 8H5 pip-H, CH2OPr, NCH2CH2), 1.31 (m5 1.2H5 pip-H), 1.03-0.98 (m, 3H, CH30Pr), 0.88-0.83 (m, 6H, CH(CHO2). ΗPLC tR = 8.5 min.
Example 157 - N-r4-Fluorobenzyl)-N-π-|'3-(4-('S)-isopropyl-2-oxo-oxazolidin-3- yl)propyπpiperidin-4-yl}-2-f4-propoxyphenv0acetarnide, oxalate d 17NLS03-O.
[0576] Prepared following the same method as described for 1 17NLS01 using N-(4-fluorobenzyl)-N-(piperidin-4-yl)-2-(4-propoxyphenyl)acetamide (104 mg, 0.27 mmol5 prepared by the procedure described for 103NLS56). Yield: 120 mg, 80%.
[0577] R/ = 0.36 (MeOHZCH2Cl2 5:95). LCMS m/z 554 [M+H]+. 1H-NMR (CDCl35 rotamers 0.6:0.4) δ 7.19-6.78 (m, 8H, Ar-H)5 4.57-4.48 (m, 0.6H5 pip-H), 4.48 and 4.42 (2s, 2H, benzyl-H), 4.18-4.12 (m5 IH5 oxa-CH2), 4.04-4.00 (m, IH5 oxa-CH2), 3.91-3.85 (m, 2H, OCH20Pr), 3.75-3.66 (m, 2.2H5 pip-H, oxa-NCH, benzyl-H), 3.49-3.43 (m, 2.2H, benzyl-H, OCONCH2), 2.98-2.80 (m, 3H, pip-H, OCONCH2), 2.33-2.25 (m, 2H3 NCH2), 2.05-1.50 (m, 10.2H3 CH(CH3)2, NCH2CH2, pip-Η, CH20Pr), 1-27 (m, 0.8H5 pip-H), 1.18-0.98 (m, 3H, CH30Pr), 0.87-0.81 (m, 6H3 CH(CHs)2). HPLC tR = 8.3 min.
Example 158 - N-a-r2-n.3-Dioxan-2-vnethvnpiperidin-4-vU-N-f4-fluorobenzyl)-2-f4- isobutoxyphenyDacetamide. oxalate (103NLSoS-F-).
[0578] Prepared following the same method as described for 117NLS01 using and 103NLS56 (262 mg, 0.657 mmol) and 2-(2-bromoethyl)-l,3-dioxane as the alkylating agent. No sodium iodide was required. Yield: 152 mg, 45%.
10579] R/ = 0.35 (MeOH/CH2Cl2 1 :9). LCMS m/z 513 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.26-6.80 (in, 8H, Ar-H), 4.63-4.39 (m, 3.6H, pip-H, dioxane-H, benzyl-H), 4.09-4.01 (m, 2H, dioxane-H), 3.78-3.64 (m, 5.2H3 pip-H, dioxane-H, CH2OiBu, benzyl-H), 3.50 (s, 1.2H, benzyl-H), 2.92-2.79 (m, 2H, pip-H), 2.43- 2.34 (m, 2H, NCH2), 2.10-1.96 (m, 3.2H, dioxane-H, pip-H, CHOiBu), 1.88-1.48 (m, 6H, pip-H, NCH2CH2), 1.35-1.24 (m, 1.8H, dioxane-H, pip-H), 1.01 (m, 6H, CH3OiBu). HPLC ?R = 8.8 min.
Example 159 - N-π-r2-O3-Dioxan-2-yl)ethyl1piperidin-4-vU-N-r2-f4- fluorophenyl)ethyl]-2-f4-isobutoxyphenyl)acetamide, oxalate (117NLS03-D).
[0580] Prepared following the same method as described for 117NLS 03 -A using 2-(2-bromoethyl)-l,3-dioxane as the alkylating agent. No sodium iodide was required. Yield: 99 mg, 70%.
[0581] R/ = 0.35 (MeOH/CH2CI2 5:95). LCMS m/z 527 [M+H]+. 1H-NMR (CDCl3, rotamers 0.7:0.3) δ 7.18-6.80 (m, 8H, Ar-H), 4.58-4.54 (m, IH, dioxane-H), 4.48-4.41 (m, 0.3H, pip-H), 4.10-4.06 (m, 2H, dioxane-H), 3.77-3.66 (m, 5.4H, dioxane- H, benzyl-H, CH2OiBu)9 3.64-3.52 (m, 1.3H, benzyl-H, pip-H), 3.37-3.32 (m, 2H, CH2NCO), 2.99 and 2.89 (2m, 2H, pip-H), 2.82-2.76 (m, 2H3 ArCH2), 2.49-2.39 (m, 2H, NCH2), 2.12-2.00 (m, 2.6H, dioxane-H, pip-H, CHoiBu), 1.88-1.67 (m, 6H, pip-H, CH2OiBu, NCH2CH2), 1.35-1.31 (m, 2.4Η, dioxane-H, pip-H), 1.00 (t, 6H, J = 6.6, CH3OiBu)- HPLC to = 8.8 min.
Example 160 - N-H-^-0 ,3-Dioxan-2-vnethyl1piperidin-4-vU-N-r2-f 4- fluorophenyl)ethyl1-2-(4-propoxyphenyl)acetamide, oxalate (117NLS03-E). [0582] Prepared following the same method as described for 117NLS03-B using 2-(2-bromoethyl)-l,3-dioxane as the alkylating agent. No sodium iodide was required. Yield: 90 mg, 65%.
[0583] R/ = 0.23 (MeOH/CH2Cl2 5:95). LCMS m/z 513 [M+H]+. 1H-NMR (CDCl3, rotamers 0.7:0.3) δ 7.21-6.81 (m, 8H5 Ar-H)5 4.58-4.54 (m, IH5 dioxane-H), 4.48-4.42 (m 0.3H5 pip-H), 4.10-4.06 (m5 2H5 dioxane-H)5 3.91-3.86 (m, 2H5 CH20Pr), 3.77-3.69 (m, 3.4H, dioxane-H, benzyl-H)5 3.63-3.56 (m, 1.3H5 benzyl-H5 pip-H), 3.38- 3.31 (m, 2H, CH2NCO), 2.99 and 2.89 (2m, 2H5 pip-H), 2.82-2.76 (m, 2H, ArCH2), 2.49- 2.39 (m, 2H5 NCH2), 2.12-2.00 (m, 1.6H5 dioxane-H, pip-H), 1.87-1.65 (m, 8H, pip-H, CH20Pr, NCH2CH2), 1.35-1.31 (m5 2.4Η, dioxane-H, pip-H), 1.05-1.00 (m5 3H, CH30Pr). HPLC /R = 8.0 min.
Example 161 - N-n-r2-ri.3-Dioxan-2-vnethyllpiperidin-4-vU-N-(4-fluorobenzvn-2-(4- propoxyphenvDacetamide. tartrate (117NLS03-F").
[0584] Prepared following the same method as described for 117NLS03-C using 2-(2-bromoethyl)-l,3-dioxane as the alkylating agent. No sodium iodide was required. Yield: 107 mg5 79%.
[0585] R/ = 0.41 (MeOH/CH2Cl2 5:95). LCMS m/z 499 [M+H]+. 1H-NMR (CDCl35 rotamers 0.6:0.4) δ 7.20-6.80 (m5 8H, Ar-H)5 4.62-4.56 (m, 0.6H5 pip-H), 4.54- 4.51 (m, IH5 dioxane-H), 4.49 and 4.43 (2s, 2H5 benzyl-H)5 4.08-4.04 (m, 2H5 dioxane- H), 3.92-3.87 (m, 2H, OCH20Pr), 3.76-3.68 (m, 3.2H, pip-H, dioxane-H, benzyl-H), 3.50 (s, 1.2H, benzyl-H), 2.90-2.83 (m, 2H, pip-H), 2.43-2.36 (m, 2H5 NCH2), 2.10-1.98 (m, 2.2H, dioxane-H, pip-H5), 1.86-1.51 (m, 8H5 pip-H5 CH2OPr5 NCH2CH2), 1.32-1.27 (m, 1.8H5 dioxane-H5 pip-H), 1.05-0.99 (m, 3H, CH3). HPLC tR = 7.6 min.
Example 162 - N-(l-r2-π.3-Dioxan-2-vπethyllpiρeridin-4-vU-N-('4-fluorobenzvπ-N'-('4- isobutoxybenzvDcarbamide, tartrate (117NLS25).
[0586] Prepared following the same method as described for 117NLS 01 using 2-(2-bromoethyl)-l53-dioxane (24 μL, 0.18 mmol) as the alkylating agent and N-(4- fluoroben2yl)-N'-(4-isobutoxyben2yl)-N-(piperidin-4-yl)carbamide (76ELH18, 50 mg5 0.12 mmol). No sodium iodide was required. Yield: 38 mg, 60%. [0587] R/ = 0.32 (MeOH/CH2Cl2 1:9). LCMS m/z 528 [M+H]+. 1H-NMR (CDCl3) δ 7.18-6.74 (m, 8H, Ar-H)5 4.53 (t, IH, J= 5.1, dioxane-H), 4.46 (t, IH, J= 5.3, NH), 4.33-4.25 (m, 5H, pip-H, benzyl-H), 4.08-4.04 (m, 2H, dioxane-H), 3.75-3.68 (m, 2H, dioxane-H), 3.66 (d, 2H, J = 6.6, CH2OiBu), 2.93-2.88 (m, 2H, pip-H), 2.43-2.39 (m, 2H, NCH2), 2.09-1.98 (m, 4H, CHOiBu, dioxane-H, pip-H), 1.77-1.56 (m, 6H, pip-H, NCH2CZZ2), 1.32-1.28 (m, IH, dioxane-H), 0.99 (d, 6H3 J= 6.6, CH3OiBu)- HPLC tR = 8.7 min.
Example 163 - N-(l-r2-π,3-Dioxan-2-yl)ethyllpiperidin-4-yl>-N-r4-fluorobenzylV2-(4- fluorophenvDacetamide, tartrate (117NLS 87- A).
[0588] To a solution of 118AF52-95 (300 mg, 0.93 mmol) and triethylamine (0.52 mL, 3.72 mmol) in dry THF (10 mL) at 00C a solution of 4-fluoroρhenylacetyl chloride (0.19 mL, 1.39 mmol) in THF (5 mL) was added dropwise and stirring was continued at rt for 3 h. The reaction mixture was filtered and the filtrate evaporated to dryness. The residue was partitioned between ethyl acetate and IM NaOH, the organic layer washed with brine, dried over Na2SO4, filtered and evaporated. Purification by silica gel column chromatography, eluting with a stepwise gradient of 0-8% methanol in dichloromethane, followed by purification of the compound by passage over an acidic ion exchange SPE cartridge, afforded the desired compound (131 mg, 31%), which was converted to its tartrate form as described above.
[0589] R/ = 0.39 (MeOH/CH2Cl2 1 :9). LCMS m/z 459 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.25-6.88 (m, 8H, Ar-H), 4.58-4.52 (m, 0.6H, pip-H), 4.50 (t, IH, J= 5.1, dioxane-H), 4.48 and 4.44 (2s, 2H, benzyl-H), 4.06-4.02 (m, 2H, dioxane-H), 3.78 and 3.50 (2s, 2H, benzyl-H), 3.72-3.64 (m, 2.4H, pip-H, dioxane-H), 2.84 (m, 2H5 pip-H), 2.40-2.35 (m, 2H, NCH2), 2.07-1.99 (m. 2.2H, dioxane-H, pip-H), 1.85-1.50 (m, 6H, pip-H, NCH2CH2), 1.30-1.25 (m, 1.8H, dioxane-H, pip-H). HPLC fe = 6.9 min.
Example 164 - N-π-r2-π.3-Dioxan-2-vπethyllpiperidin-4-vU-N-r4-fluorobenzyl')-2-p- tolylacetamide. tartrate (117NLS87-B~).
[0590] Prepared following the same method as described for 117NLS87-A using 4-methylphenylacetyl chloride and 118AF52-95 (300 mg, 0.93 mmol). Yield: 119 mg, 28%. [0591] R/ = 0.43 (MeOH/CH2Cl2 1:9). LCMS m/z 455 [M+H]+. 1H-NMR (CDCl3, rotamers 0.5:0.5) δ 7.17-6.87 (m, 8H5 Ar-H), 4.60-4.53 (m, 0.5H, pip-H), 4.50 (t5 IH, J= 5.1, dioxane-H), 4.48 and 4.41 (2s, 2H, benzyl-H), 4.05-4.01 (m, 2H5 dioxane-H), 3.77-3.66 (m, 3.5H5 pip-H, benzyl-H, dioxane-H), 3.50 (s, IH, benzyl-H), 2.87-2.80 (m5 2H, pip-H), 2.40-2.34 (m, 2H, NCH2), 2.30 and 2.28 (2s, 3H, CH3), 2.07-1.95 (m, 2H5 dioxane-H, pip-H), 1.83-1.50 (m, 6H, pip-H, NCH2CH2), 1.29-1.25 (m, 2H, dioxane-H, pip-H). HPLC tR = 7.7 min.
Example 165 - 2-Benzoiuran-5-yl-N-n-r2-('1.3-dioxan-2-yl)ethvnpiperidin-4-vU-N-r4- fluorobenzvnacetamide. tartrate O28NLS22-A).
[0592] Benzofuran-5-yl-acetic acid was prepared adapting a procedure by Dunn et al. (J. Med. Chem., 1986, 29, 2326) and converted into the corresponding acetyl chloride by treatment with oxalylchloride. The title compound was prepared from 118AF52-95 (58 mg, 0.18 mmol) following the same method as described for 117NLS87- A. Yield: 27 mg, 43%.
[0593] R/ = 0.52 (MeOH/CH2Cl2 1:9). LCMS m/z 481 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.64-6.68 (m, 9H, Ar-H)5 4.62-4.54 (m, 0.6H, pip-H) 4.53- 4.44 (m, 3H5 dioxane-H. benzyl-H)5 4.07-4.03 (m, 2H5 dioxane-H), 3.82-3.61 (m, 3.2H5 pip-H, benzyl-H, dioxane-H), 3.45 (s, 1.2H5 benzyl-H), 2.91-2.80 (m, 2H, pip-H), 2.44- 2.35 (m, 2H5 NCH2), 2.08-1.98 (m, 2.2H, dioxane-H, pip-H), 1.85-1.56 (m, 6H, pip-H, NCH2CH2), 1.32-1.27 (m, 1.8Η, dioxane-H, pip-H). HPLC tR = 6.6 min.
Example 166 - 2-f2.3-Dihvdrobenzofuran-5-yl)-N-π-r2-π,3-dioxan-2-yl)ethyl1piperidin- 4-vU-N-(4-fluorobenzyl)acetamide, tartrate (128NLS22-B).
[0594] The compound (2,3-Dihydrobenzofuran-5-yl)acetic acid was prepared adapting a procedure by Dunn et al. (J. Med. Chem., 1986, 29, 2326) and converted into the corresponding acetyl chloride by treatment with oxalylchloride. The title compound was prepared from 118AF52-95 (58 mg, 0.18 mmol) following the same method as described for 117NLS87-A. Yield: 27 mg, 31%.
[0595] R/ = 0.50 (MeOH/CH2Cl2 1 :9). LCMS m/z 483 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.10-6.60 (m, 7H, Ar-H), 4.55-4.40 (m, 5.6H, pip-H, dioxane-H. benzyl-H, ArOCH2), 4.01-3.97 (m, 2H, dioxane-H), 3.72-3.62 (m, 3.2H, pip- H, benzyl-H, dioxane-H), 3.41 (s, 1.2H5 benzyl-H), 3.14-3.06 (m, 2H, OCH2CH2), 2.80 (m, 2H, pip-H), 2.35-2.30 (m, 2H, NCH2), 1.99-1.93 (m, 2.2H, dioxane-H, pip-H), 1.80- 1.44 (m, 6H5 pip-H, NCH2CH2), 1.27-1.22 (m, 1.8Η, dioxane-H, pip-H). HPLC tR = 6.9 min.
Example 167 - N-π-r2-(2.2-Dimethyl-1.3-dioxolan-4-vnethvnpiperidin-4-vU-N-f4- fluorobenzyl')-2-f4-isobutoxyphenyl')acetamide. tartrate O 17NLS37).
[0596] l-(2l,2'-Dimethyl-r,3'-dioxolan-4'-yl)ethanol was prepared according to literature procedures (Carman R.M et al., Aust. J. Chem., 1998, 57, 955) and oxidized to the aldehyde by treatment with pyridinium chlorochromate. The crude aldehyde (80 mg, 0.55 mmol) was added to a solution of 103NLS56 (184 mg, 0.46 mmol) in methanol (5 mL). Acetic acid (0.05 mL) was added, followed by sodium cyanoborohydride (58 mg, 0.92 mmol) and the mixture stirred overnight at it. The solvent was removed and the residue partitioned between dichloromethane and IM NaOH. The organic layer was washed with sat. NH4CI, dried over Na2SO4, filtered and evaporated. Purification by silica gel column chromatography eluting with 0-5% methanol in dichloromethane afforded the desired compound (50 mg, 21%), which was converted into its tartrate salt.
[0597] R/ = 0.39 (MeOH/CH2Cl2 1:9). LCMS m/z 527 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.22-6.79 (m, 8H, Ar-H), 4.62-4.54 (m, 0.6H, pip-H), 4.49 and 4.42 (2s, 2H, benzyl-H), 4.06-3.98 (m, IH, dioxolane-H), 3.75-3.66 (m,4.4H, pip-H, CH20JBu5 benzyl-H), 3.48 (m, 2H, dioxolane-H), 2.89-2.83 (m, 2H, pip-H), 2.45-2.25 (m, 2H, NCH2), 2.07-1.99 (m, 2.2H, pip-H, CHOiBu), 1.85-1.51 (m, 6H, pip-H, NCH2CH2), 1.36-1.28 (m, 6.8Η, C(CH3)2, pip-H), 1.02-0.99 (m, 6H5 CH3OiBu)- HPLC tR = 9.3 min.
Example 168 - 4-r2-fTosyloxy)ethyll-1.3-dioxane f 128NLS46-B\
[0598] A suspension of 1,3,5-pentanetriol (1.01 g, 8.33 mmol), paraformaldehyde (0.46 g) and methanesulfonic acid (0.33 mL) in DMF (3 mL) is heated for 10 min at 13O0C under microwave irradiation. The mixture was partitioned between ethyl acetate and water, the organic layer dried over Na2SO4, filtered and evaporated. The residue was dissolved in methanol (3 mL), cone. HCl (0.09 mL) added, and the mixture heated at 8O0C for 10 min under microwave irradiation. Ethyl acetate and 2M NaOH were added, the aqueous layer extracted twice with ethyl acetate and the combined organic layers washed with brine, dried over Na2SO4, filtered and evaporated. The crude product was treated with jo-tosylchloride and DMAP following literature procedures (Moune et ah, J. Org. Chem., 1997, 62, 3332). The title compound (1.18 g, 49% overall crude yield) was obtained as a yellowish oil, which was used without purification.
Example 169 - N-(l-[2-π,3-Dioxan-4-yl')ethyllpiperidin-4-yU-N-r4-fluorobenzyl')amine C128NLS52V
[0599] To a suspension of 4-piperidone monohydrate hydrochloride (1.26 g, 8.23 mmol) in acetonitrile (100 mL), potassium carbonate (3.4 g, 24.6 mmol) was added, followed by the tosylate 128NLS46-B (3.54 g, 12.36 mmol) and sodium iodide (1.85 g, 12.35 mmol) and stirring was continued overnight at 6O0C. The mixture was filtered, the filtrate evaporated in vacuo and the residue partitioned between IM NaOH and ethyl acetate. The organic layer was separated, the aqueous layer extracted twice with ethyl acetate and the combined organic layers dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with a stepwise gradient of 0-4% methanol in dichloromethane, afforded l-[2- (l ,3-dioxan-4-yl)ethyl]piperidin-4-one (128NLS50, 1.73 g, 98%).
[0600] To a solution of 128NLS50 (1.73 g, 8.13 mmol) in methanol (100 mL) was added dropwise 4-fluorobenzylamine (0.93 mL, 8.13 mmol) and acetic acid. Sodium cyanoborohydride (2.15 g, 40 mmol) was added slowly to the mixture at O0C and stirring was continued at rt overnight. The reaction mixture was concentrated in vacuo and the residue partitioned between dichloromethane and IM NaOH, the aqueous layer extracted twice with dichloromethane and the combined organic layers dried over Na2SO4, filtered and evaporated to dryness. Purification of the residue by a short silica gel column chromatography eluting with 0-30% methanol in dichloromethane gave the title compound (1.51 g, 58%) as a colorless solid.
Example 170 - N-(l-r2-π.3-Dioxan-4-vnethvnpiperidin-4-vn-N-r4-fluorobenzylV2-r4- isobutoxyphenvDacetamide. tartrate f 128NLS62).
[0601] Prepared following the same method as described for 117NLS87-A using 4-isobutoxyphenylacetyl chloride and 128NLS52 (480 mg, 1.49 mmol). Yield: 458 mg, 60%.
[0602] Rf = 0.36 (MeOH/CH2Cl2 1:9). LCMS m/z 513 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.21-6.80 (m, 8H, Ar-H), 5.01 (d, IH5 J= 6.1, dioxane-H), 4.66-4.56 (m, 1.6H, pip-H, dioxane-H) 4.51 and 4.44 (2s, 2H, benzyl-H), 4.09-4.05 (m, IH, dioxane-H), 3.77 and 3.51 (2s, 2H, benzyWi), 3.70-3.57 (m, 4.4H, ρiρ-H, dioxane-H, CH2OiBuX 2.91-2.83 (m, 2H, pip-H), 2.45-2.34 (m, 2H, NCH2), 2.10-2.00 (ra, 2.2H, pip-H, CHoiBu), 1.85-1.26 (m, 8.8H5 pip-H, dioxane-H, NCH2CH2), 1.03-1.00 (m, 6H, CH3OiBu). HPLC /R = 8.8 min.
Example 171 - N-(l-r2-(L3-Dioxan-4-yl')ethvnpiperidin-4-vU-N-(4-fluorobenzvn-2-r4- trifluoromethylphenvOacetamide. tartrate (128NLS54-A*).
[0603] Prepared following the same method as described for 117NLS 87- A using 4-trifluorophenylacetyl chloride and 128NLS52 (1 16 mg, 0.32 mmol). Yield: 52 mg, 32%.
[0604] R/ = 0.42 (MeOH/CH2Cl2 1:9). LCMS m/z 509 [M+H]+. 1H-NMR (CDCl3, retainers 0.6:0.4) δ 7.60-6.90 (m, 8H, Ar-H), 4.99 (d, IH, J = 6.1, dioxane-H), 4.65-4.54 (m, 1.6H, pip-H, dioxane-H), 4.52 and 4.47 (2s, 2H, benzyl-H), 4.07-4.04 (m, IH, dioxane-H), 3.88 (s, 0.8H, benzyl-H), 3.69-3.56 (m, 3.6H5 benzyl-H, pip-H, dioxane- H), 2.89 (m, 2H, pip-H), 2.49-2.31 (m, 2H, NCH2), 2.07-1.99 (m, 1.2H, pip-H), 1.89-1.36 (m, 8.8H, pip-H, dioxane-H, NCH2CH2). ΗPLC /R = 7.3 min.
Example 172 - 2-f4-CvanophenylVN-π-r2-ri.3-dioxan-4-vnethvnpiperidin-4-yl>-N-f4- fluorobenzvnacetamide, tartrate d28NLS54-O.
[0605] 4-Cyanophenylacetic acid was synthesized according a method by Jaeger et al. (J. Chem. Soc, 1941, 744-747) and converted to the corresponding acetyl chloride by treatment with oxalylchloride. The title compound was prepared following the same method as described for 117NLS87-A using 4-cyanophenylacetyl chloride and 128NLS52 (116 mg, 0.32 mmol). Yield: 60 mg, 40%.
[0606] Rf = 0.40 (MeOΗ/CΗ2Cl2 1:9). LCMS m/z 466 [M+H]+. 1H-NMR (CDCl3, rotamers 0.7:0.3) δ 7.62-6.89 (m, 8H, Ar-H), 4.97 (d, IH, J = 6.1, dioxane-H), 4.63 (m, IH, dioxane-H), 4.59-4.47 (m, 2.7H, pip-H, benzyl-H), 4.06-4.02 (m, IH, dioxane-H), 3.86 (s, 0.6H, benzyl-H), 3.69-3.55 (m, 3.7H, benzyl-H, pip-H, dioxane-H), 2.91-2.86 (m, 2H, pip-H), 2.47-2.30 (m, 2H, NCH2), 2.05-1.39 (m, 1OH, pip-H, dioxane- H, NCH2CH2). ΗPLC tR = 4.3 min. Example 173 - N-(4-FluorobenzylV2-f4-isobutoxyphenyl)-N-π-r2-(2-oxo-imidazolidin- l-yl)ethyllpiperidin-4-vUacetamide. hydrochloride (69NLS971
[0607] Prepared following the same method as described for 117NLS01 using 103NLS56 (240 mg, 0.60 mmol) and l-(2-tosyloxyethyl)-2-imidazolidinone as the alkylating agent. Yield: 95 mg, 31%.
[0608] LCMS m/z 511 [M+H]+.1H-NMR (CD3OD, rotamers 0.6:0.4) δ 7.24- 6.81 (m, 8H, Ar-H), 4.56 and 4.52 (2s, 2H, benzyl-H), 4.41-4.37 and 3.93-3.88 (m, IH, pip-H), 3.84 and 3.56 (2s, 2H, benzyl-H), 3.73-3.69 (m, 2H, CH2OiBu), 3.46-3.20 (m, 6H, imid-CH2, NCH2CH2), 2.99-2.85 (m, 2H, pip-H), 2.44 (m, 2H, NCH2), 2.10-1.96 (m, 3.2H, pip-H, CHoiBu), 1.67-1.62 (m, 3H, pip-H), 1.30 (m, 0.8H, pip-H), 1.03-0.99 (m, 6H, J= 6.6, CH3OiBu)-HPLC tR = 9.5 min.
[0609] Choosing the appropriate secondary amines (prepared in analogy to the method described for 103NLS56), following compounds were prepared using a similar procedure:
Example 174 - 2-(4-MethoxyphenylVN-r4-methylbenzvπ-N-(l-|"2-(2-oxo-imidazolidin-l- yl)ethyl]piperidin-4-yl)acetamide, hydrochloride (63ELH39-B).
[0610] LCMS m/z 465 [M+H]+.Η-NMR (CDCl3, rotamers 0.6:0.4) δ 7.30- 6.80 (m, 8H), 4.60-4.53 (m, 0.6H), 4.50 and 4.43 (2s, 2H), 3.78 (m, 4.2H), 3.51 (s, 1.2H), 3.46-3.24 (m, 6H), 2.92-2.79 (m, 2H), 2.46-2.40 (m, 2H), 2.35 and 2.29 (2s, 3H), 2.11- 2.05 (m, 1.2H), 1.92-1.86 (m, 0.8H), 1.65-1.50 (m, 3.2H, partly covered by HDO signal), 1.31 (m, 0.8H).
Example 175 - N-(4-Fluorobenzyl)-2-('4-isopropoxyphenylVN-(l-r3-C3-methyl-2-oxo-
2.3-dihvdro-benzoimidazol-l-yl)propyl]piperidin-4-vUacetarnide: hydrochloride
(103NLSSgI
[0611] Prepared following the same method as described for 117NLS01 using N-(4-fluorobenzyl)-N-(piperidin-4-yl)-2-(4-isoρroxyphenyl)acetamide (229 mg, 0.59 mmol) and l-(3-chloropropyl)-3-methyl-l,3-dihydrobenzimidazol-2-one as the alkylating agent.
[0612] Yield: 205 mg, 61%. R7= 0.29 (MeOH/CH2Cl2 5:95). LCMS m/z 573 [M+H]+. 1H-NMR (CDCl3, rotamers 0.5:0.5) δ 7.18-6.78 (m, 12H, Ar-H), 4.59-4.43 (m, 3.5H, pip-H, OCH, benzyl-H), 3.88 (t, 2H, J = 6.8, NCONCH2), 3.74 (m, 1.5H, pip-H, benzyl-H), 3.49 (s, IH, benzyl-H), 3.38 and 3.37 (2s, 3H5 NCH3), 2.93-2.79 (m, 2H, pip- H), 2.36-2.29 (m, 2H, NCH2), 2.02-1.95 (m, IH, pip-H), 1.90-1.46 (m, 6H, pip-H, NCH2CH2), 1.31-1.25 (m, 7Η, pip-H, CH(CH3)2). ΗPLC tR = 8.0 min.
[0613] Choosing the appropriate secondary amines (prepared in analogy to the method described for 103NLS56) and alkylating agents, following compounds were prepared using a similar procedure:
N- { 1 -[2-(2,4-Dioxo-l ,4-dihydro-2Η-quinazolin-3-yl)ethyl]piperidin-4-yl}-2-(4- methoxyphenyl)-N-(4-methylbenzyl)acetamide, hydrochloride (63ELH29A).
2-(4-Methoxyphenyl)-N-(4-methylbenzyl)-N-{ l-[3-(2-oxo-2,3- dihydrobenzoimidazol- 1 -yl)propyl]piperidin-4-yl} -acetamide, hydrochloride (50ELH89).
N-(4-Fluorobenzyl)-2-(4-isopropoxyphenyl)-N-{ 1 -[4-(2-oxo-2,3- dihydrobenzoimidazol-1 -yl)butyl]piperidin-4-yl} acetamide, hydrochloride (63ELH91 ).
N-{l-[2-(2,4-Dioxo-l,4-dihydro-2H-quinazolin-3-yl)ethyl]piperidin-4-yl}-N-(4- fluorobenzyl)-2-(4-isopropoxyphenyl)acetamide, hydrochloride (63ELH89).
Example 176 - 4-(4-FluorobenzyIamino)-piρeridine-l-carboxylic acid benzyl ester α i8AF93-5n.
[0614] A solution of 4-fluorobenzylamine (5.48 g, 43.8 mmol) in a mixture of methanol and acetic acid (5:1, 60 mL) was added dropwise to a solution of benzyl 4-oxo- 1-piperidine carboxylate (10.2 g, 43.8 mmol) in methanol (150 mL) at rt. To this mixture sodium cyanoborohydride (5.50 g, 87.5 mmol) was slowly added. After 20 hours stirring at rt the reaction mixture was neutralized and the solvent was removed by evaporation under reduced pressure. The residue was partitioned between dichloromethane and water. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with 7% methanol in dichloromethane, afforded the desired compound (9.0 g, 60 %).
[0615] R/ = 0.56 (MeOH/CH2Cl2 5:95). LCMS m/z 343 [M+H]+. HPLC /R = 6.2 min.
Example 177 - N-π-Benzyloxycarbonylpiperidin-4-viyN-(4-fluorobenzvP-N'-f4- isopropoxvbenzvDcarbamide C118AF97-120). [0616] l,8-Bis(dimethylamino)-naphtalene (3.19 g, 14.9 mmol) was added to a solution of 4-(isopropoxy)phenyl acetic acid (2.89 g, 14.9 mmol) in dry tetrahydrofliran (18 mL) at rt under argon atmosphere. After 25 minutes stirring at it diphenylphosphoryl azide (4.10 g, 14.9 mmol) was added dropwise and the mixture refluxed for 6 hours. It was allowed to cool to rt and then stored at — 200C overnight to precipitate out the ammonium phosphate salt. A mixture of diethyl ether and ethyl acetate (1 : 1 v/v, 25 mL) was added to the cold reaction mixture. The precipitate was filtered from the reaction mixture and washed with diethyl ether: ethyl acetate (1:1 v/v, 20 mL). The filtrate was evaporated to dryness giving l-isocyanatomethyl-4-isopropoxybenzene as an oil (3.2 g), which was used in the next step without further purification.
[0617] Sodium carbonate (3.5 g, 25.3 mmol) was added to the solution of 4- (4-fluorobenzyl amino)-piperidine-l-carboxylic acid benzyl ester 118AF93-51 (5.7g, 16.7 mmol) in dry tetrahydroruran (20 mL). To this suspension a solution of 1- isocyanatomethyl-4-isopropoxybenzene (3.2 g, 16.7 mmol) in dry tetrahydrofliran (10 mL) was added under argon atmosphere. The reaction mixture was stirred overnight at rt. Afterwards the mixture was partitioned between dichloromethane and water. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with 8% methanol in dichloromethane afforded the desired compound (2.0 g, 22%).
[0618] R/= 0.36 (MeOH/CH2Cl2, 5:95). LCMS m/z 534 [M+H]+. HPLC fR = 10.2 min.
Example 178 - N-r4-FluorobenzylVN'-('4-isopropoxybenzvπ-N-piperidin-4-yl-carbamide. oxalate (118AF99-12n.
[0619] The desired compound was obtained by hydrogenation of 118AF97- 120 (2.0 g, 3.75 mmol) in absolute ethanol (100 mL) using palladium on carbon as a catalyst. The product was purified by column chromatography on silica gel eluting with stepwise gradient of 5-10 % methanol in dichloromethane. Yield: 1.16 g, 77 %.
[0620] R/ = 0.10 (MeOH/CH2Cl2 10:90). LCMS m/z 400 [M+H]+ 1H NMR (CDCl3) δ 7.19 (m, 2H, Ar-H), 7.01-6.69 (m, 4H, Ar-H)5 6.76 (m, 2H5 Ar-H)5 4.51-4.40 (m, 3H, pip-H, OCH(CH3), NH)5 4.35 (s, 2H, benzyl-H), 4.28 (s, IH, benzyl-H)5 4.27 (s, IH, benzyl-H), 3.14-3.07 (m, 2H, pip-H), 2.74-2.68 (m, 2H, pip-H), 2.10 (broad s, IH5 NH), 1.78-1.70 (m, 2H5 pip-H), 1.58-1.48 (m5 2H5 pip-H)5 1.31 (d, 6H, J = 6.0, OCH(CH5)). HPLC rR= 5.9 min.
Example 179 - N-(l-r2-rL3-Dioxolan-2-vnethvnpiperidin-4-yl>-N-f4-fluorobenzvn-N'- (4-isopropoxy-benzyl)carbamide, oxalate (T30AF10-147).
[0621] Potassium carbonate (0.2 Ig, 1.50 mmol) was added to a solution of 118AF99-121 (0.3 g, 0.75 mmol) in dry N,iV-dimethylformamide (2 mL). The suspension was shaken for 30 minutes at 58°C. A solution of 2-(2-bromoethyl)-l,3-dioxolane (0.163 g, 0.90 mmol) in dry i\ζN-dimethylformamide (0.4 mL) was added dropwise to the warm suspension and the heating was continued overnight. The mixture was allowed to cool to rt, then filtered and partitioned between water and dichloromethane. The organic layer was washed with a aqueous solution of 4% magnesium sulphate and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with 4% methanol in dichloromethane, afforded the desired compound (197 mg, 53 %). The product was converted to its oxalate form as described above.
[0622] R7 = 0.39 (MeOH/CH2Cl2 4:94). LCMS m/z 500 [M+H]+. 1H NMR (CDCl3) δ 7.17 (m, 2H5 Ar-H), 7.00-6.95 (m, 4H, Ar-H), 6.76 (m, 2H5 Ar-H)5 4.88 (t, IH5 J = 4.8, dioxolane-H)5 4.51-4.44 (m, 2H5 NH5 CH(CH3)2), 4.36-4.26 (m5 5H5 benzyl-H5 pip-H)5 3.95-3.80 (m, 4H, dioxolane-H)5 2.98-2.91 (m, 2H5 pip-H), 2.48-2.43 (m, 2H, NCH2), 2.10-2.01 (m5 2H5 pip-H), 1.85-1.79 (m, 2H, NCH2CH3), 1.76-1.58 (m, 4H5 pip- H)5 1.30 (d, 6H, J= 6.0, CH(CHj)2). ΗPLC tR= 6.9 min.
[0623] Choosing the appropriate secondary amines (prepared in analogy to the method described for 103NLS56), following compounds were prepared using the same procedure:
N- { 1 -[2-(1 ,3-Dioxolan-2-yl)ethyl]piperidin-4-yl]-2-(4-methoxyphenyl)-N-(4- methylbenzyl)acetamide5 hydrochloride (63ELΗ29B).
N- { 1 -[2-( 153-Dioxolan-2-yl)ethyl]piperidin-4-yl } -N-(4-fluorobenzyl)-2-(4- isobutoxyρhenyl)acetamide, hydrochloride (74AKU06-2).
N- { 1 -[2-(1 ,3-Dioxolan-2-yl)ethyl]piperidin-4-yl } -2-(4-isopropoxyphenyl)-N-(4- methylbenzyl)acetamide, hydrochloride (76ELH07).
N-{ 1 -[2-(153-Dioxolan-2-yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4- propoxyphenyl)acetamide5 tartrate (38PH50). Example 180 - N-(4-FluorobenzylVN'-f4-isopropoxybenzylVN-π-r2-('rSV4-methyl-1.3- dioxolane-Σ-vπethvnpiperidin^-vπcarbamide, oxalate C130AF12-148').
[0624] 4 M HCl (0.5 mL) and water (0.5 mL) were added to a solution of 130AF 10-147 (50 mg, 0.10 mmol) in 1.4-dioxane (1 mL). The mixture was stirred in a sealed flask for 10 minutes under microwave irradiation at 1200C. Afterwards the mixture was partitioned between dichloromethane and water. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. The residue was dissolved in 1.4- dioxane (1 mL) and a solution of (S)-(+)-propylene glycol (39 mg, 0.51 mmol) in 1.4- dioxane (0.5 mL) was added. After addition of HCl (4M in dioxane, 0.5 mL) the mixture was stirred in a sealed flask for 20 minutes under microwave irradiation at 1200C. The mixture was partitioned between saturated sodium bicarbonate solution and dichloromethane. The organic layer was evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with a stepwise gradient of 4-8 % methanol in dichloromethane afforded the desired compound (2.1 mg, 4 %). The product was converted to its oxalate form as described above.
[0625] R/ = 0.36 (MeOH/CH2Cl2 4:94). LCMS m/z 514 [M+H]+. HPLC rR = 7.2 min.
Example 181 - N-f4-Fluorobenzyl')-N'-(4-isopropoxybenzyl)-N-['l-(3-moφholin-4-yl- propyl)piperidin-4-yl1carbamide, oxalate (130AF09-145).
[0626] A solution of l-chloro-3-bromopropane in dry tetrahydrofuran (2 mL) was added to a cold suspension of morpholine (200 mg, 2.29 mmol) and sodium carbonate (0.63 g, 4.56 mmol) in dry tetrahydrofuran (8 mL) at 00C. The mixture was stirred at 450C overnight. The mixture was allowed to cool to rt, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with a mixture of ethyl acetate and w-heptane (70:30), afforded 3-chloro-l-morphoIin-4-yl- propane (156 mg, 42 %).
[0627] A solution of 3-chloro-l-morpholin-4-yl-propane (7.6 mg, 0.046 mmol) in dry iVVV-dimethylformamide (0.10 mL) was added to a solution of 1 18AF99- 121 (15 mg, 0.037 mmol) and caesium carbonate (40 mg, 0.123 mmol) in a mixture of dry JV^-dimethylformamide and acetonitrile (1:2, 0.30 mL). After addition of sodium iodide (7.0 mg, 0.047 mmol) the mixture was shaken overnight at 600C. The mixture was allowed to cool to rt. Acetonitrile was removed by evaporation under reduced pressure and the residue was partitioned between dichloromethane (2 mL) and water (1 mL). The organic layer was evaporated to dryness. Purification of the residue by preparative reversed phase HPLC (Ci8) afforded the desired compound (6.1 mg, 32 %).
[0628] LCMS m/z 527 [M+H]+. HPLC tR = 6.2 min.
[0629] Choosing the appropriate secondary amines (prepared in analogy to the method described for 103NLS56), following compounds were prepared using a simkilar procedure:
2-(4-Methoxyphenyl)-//-(4-methylbenzyl)-iV- [ 1 -(2-morpholin-4-ylethyl)piperidin- 4-yl]acetamide, dihydrochloride (63ELH40-2).
2-(4-Methoxyphenyl)-N-(4-methylbenzyl)-N- [ 1 -(3 -morpholin-4- ylpropyl)piperidin-4-yl]acetamide, dihydrochloride (63ELH41-2).
N-(4-Fluoroberrzyl)-2-(4-isobutoxyphenyl)-JV- [ 1 -(3 -morpholin-4- ylpropyl)piperidin-4-yl]acetamide, dihydrochloride (74AKU07-2). iV-(4-Fluoroben2yl)-2-(4-isopropoxyphenyl)-iV-[ 1 -(3-morpholin-4-yl- propyl)piperidin-4-yl]acetamide, dihydrochloride (76ELH14-A).
Example 182 - N-f4-Fluorobenzyl')-N'-('4-isopropoxybenzyl)-N-['l-('3-piperidin-l-yl- propyl)piperidin-4-yl]carbamide, oxalate (130AF09-146).
[0630] The desired compound was synthesized from piperidine, l-chloro-3- bromopropane and 118AF99-121 (15 mg, 0.037 mmol) using the same method as for preparation of 130AF09-145. Yield: 5.8 mg, 30 %.
[0631] LCMS m/z 525 [M+H]+. HPLC tR = 6.8 min.
Example 183 - N-(4-Fluorobenzyl')-Ny-(4-isopropoxybenzvD-N-ri-(3-r(S')-4-isopropyl-2- oxazolidinon- 1 -yl-propyDpiperidin-4-yl'lcarbamide, tartrate Cl 3 OAF 14-152).
[0632] The desired compound was synthesized from (4<S)-3-(3-chloropropyl)- 4-isoρropyloxazolidinon-2-one 103NLS94 (7.4 mg, 0.045 mmol) and 118AF99-121 (15 mg, 0.037 mmol) using the same method as for preparation of 130AF09-145. Yield: 3.3 mg, 16%.
[0633] LCMS m/z 569 [M+H]+. HPLC tR = 8.2 min. Example 184 - N-r4-FluorobenzylVN'-f4-isopropoxybenzylVN-ll-r2-(2.5.5-trimethyl- 1.3-dioxan-2-yr)ethvn )piperidin-4-vncarbamide. oxalate Cl 30AF07- 143V
[0634] The desired compound was synthesized from 2-bromo- 1 -(2,5,5- trimethyl-l,3-dioxan-2-yl)-ethane (10.7 mg, 0.045 mmol) and 118AF99-121 (15 mg, 0.037 mmol) using the same method as for preparation of 130AF09-145. Yield: 8.3 mg, 15 %.
[0635] LCMS m/z 556 [M+H]+. HPLC tR = 9.6 min.
Example 185 - N-π-rS-πj-Dioxolan-Σ-vDpropyllpiperidin^-vU-N-^-fluorobenzylVN^ (4-isopropoxybenzyl)carbamide, oxalate O30AF07-131).
[0636] The desired compound was synthesized from 3-chloro-l-(l,3-dioxolan- 2-yl)-propane (6.79 mg, 0.045 mmol) and 118AF99-121 (15 mg, 0.037 mmol) using the same method as for preparation of 130AF09-145. Yield: 5.6 mg, 11 %.
[0637] LCMS m/z 514 [M+H]+. HPLC tR = 8.3 min.
Example 186 - N-[l-(2,2-Dimethyl-1.3-dioxan-5-yl)piperidin-4-yl1-N-(4-fluorobenzyIV N'-(4-isopropoxybenzyDcarbamide. oxalate d30AF05-129).
[0638] A solution of 2,2-dimethyl-l,3-dioxan-5-one (9.75 mg, 0.075 mmol) in methanol (0.10 mL) was added to a solution of 118AF99-121 (15 mg, 0.037 mmol) in methanol (0.10 mL). The reaction mixture was stirred at rt after addition of acetic acid (60 μL of 1 M solution in methanol). After 2 h stirring a solution of sodium cyanoborohydride (5 mg, 0.079 mmol) in methanol (0.10 mL) was added and stirring was continued overnight at rt. The solvent was removed by evaporation under reduced pressure and the residue partitioned between 2 M aq. sodium hydroxide and dichloromethane. The layers were separated by filtration over PTFE filter. The organic layer was evaporated to dryness. Purification of the residue by preparative reversed phase HPLC (Ci s) afforded the desired compound (2.3 mg, 12 %).
[0639J LCMS m/z 514 [M+H]+. HPLC tR = 9.0 min.
Example 187 - N-r4-Fluorobenzyl)-N'-(4-isopropoxybenzyl')-N-<r2-fl-methyl pyrrolidin- 2-vnethvn-piperidin-4-vUcarbamide. oxalate (130AF07-135^
[0640] The desired compound was synthesized from 2-(2-chloroethyl)-l- methylpyrrolidine hydrochloride (7.7 mg, 0.041 mmol) and 118AF99-121 (15 mg, 0.037 mmol) using the same method as for the preparation of 130AF09-145. Yield: 4.4 mg, 23 %.
[0641] LCMS m/z 511 [M+H]+. HPLC tR = 7.0 min.
Example 188 - N-ri-f2,2-Dimethyl-1.3-dioxan-5-vnpiρeridin-4-vn-N-('4-fluorobenzvπ-2- (4-isobutoxyphenyDacetamide, oxalate (130AF22-105).
[0642] A solution of 2,2-dimethyl-l,3-dioxan-5-one (81 mg, 0.62 mmol) in methanol (10 mL) was added dropwise to a solution of 103NLS56 (179 mg, 0.45 mmol) in methanol (10 mL). The reaction mixture was stirred at rt after addition of acetic acid (200 μL). After 2 hours sodium cyanoborohydride (56 mg, 0.90 mmol) was slowly added and stirring was continued overnight at rt. The mixture was neutralized with few drops of 2 M aq sodium hydroxide. The solvent was removed by evaporation under reduced pressure and the residue partitioned between water and dichloromethane. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with 6% methanol in dichloromethane, afforded the desired compound (98 mg, 43%).
[0643] R7 = 0.32 (MeOH/CH2Cl2, 6:94). LCMS m/z 513 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.26-6.79 (m, 8H, Ar-H), 4.63-4.54 (m, 0.6H, pip-H), 4.50 & 4.43 (2s, 2H5 benzyl-H), 3.91 & 3.88 (2d, IH, J = 5.6, dioxane-H), 3.79-3.67 (m, 6.2 H, dioxane-H, benzyl-H, pip-H, CH2OiBu), 3.51 (s, 1.2H, benzyl-H), 2.98-2.88 (m, 2H, pip- H), 2.64-2.52 (m, IH, dioxane-H), 2.38-2.28 (m, 1.2H, pip-H), 2.17-2.00 (m, 1.8H, CH(CH3)2, pip-H), 1.72-1.47 (m, 3.2H, pip-H), 1.43 (m, 0.8H, pip-H), 1.38-1.22 (m, 6H, dioxane-CHa), 1.01 (m, 6H, CH(CHa)2). HPLC tR= 10.0 min.
Example 189 - N-[I-H .3-Dioxan-5-vn-piperidin-4-ylVN-('4-fluorobenzyl)-2-f4- isobutoxyphenvDacetamide, tartrate (130AF26-164).
[0644] 3 M aq HCl (1 mL) and water (1 mL) were added to a solution of 130AF22-105 (98.2 mg, 0.19 mmol) in 1.4-dioxane (2 mL) and the mixture stirred in a sealed flask under microwave irradiation for 10 minutes at 1200C. The mixture was partitioned between water and dichloromethane and the organic layer dried over sodium sulphate, filtered and evaporated to dryness. The residue was dissolved in 1.4-dioxane (2 mL). To this solution a solution of formaldehyde (37% water solution, 101 mg, 1.16 mmol) in 1.4-dioxane (0.5 mL) was added. The reaction mixture was stirred in a sealed flask for 30 minutes under microwave irradiation at 1200C. Molecular sieves (4A) were added to the reaction mixture at rt and removed after 24 hours. The mixture was heated for an additional 20 minutes at 1200C under microwave irradiation and partitioned between dichloromethane and sodium bicarbonate. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with 6% methanol in dichloromethane, afforded the desired compound (17 mg, 18%). The product was converted to its tartrate form as described above.
[0645] R/ = 0.30 (MeOH/CH2Cl2, 6:94). LCMS m/z 485 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.21-6.80 (m, 8H, Ar-H), 4.88 (m, IH, dioxane-H), 4.61-4.56 (m, 1.6H5 dioxane-H, ρiρ-H), 4.50 & 4.43 (2s, 2H, benzyl-H), 4.12-4.06 (m, 2H, dioxane- H) 3.85-3.60 (m, 5.2 H5 dioxane-H, benzyl-H, pip-H, CH2OiBu), 3.51 (s, 1.2H, benzyl-H), 2.94-2.86 (m, 2H5 pip-H), 2.59-2.48 (m, IH5 dioxane-H), 2.37-2.28 (m, 1.2H, pip-H)5 2.17-2.01 (m, 1.8H, CH(CH3)2, pip-H), 1.68-1.46 (m, 3.2H5 pip-H), 1.46-1.30 (m, 0.8H5 pip-H), 1.02 (m, 6H, CH(CH3)2). HPLC tR= 9.5 min.
Example 190 - N-ri-(2.2-Dimethyl-1.3-dioxan-5-yl)piperidin-4-yll-N-(4-fluorobenzvn-2- (4-fluorophenyl)acetamide, tartrate (130AF35-168).
[0646] The desired compound was synthesized from 2,2-dimethyl-l,3-dioxan- 5-one (59 mg, 0.45 mmol) and N-(4-fluorobenzyl)-2-(4-fluorophenyl)-N-piperidin-4-yl- acetamide (83 mg, 0.24 mmol) using the same method as for preparation of 130AF22- 105. The starting material iV-(4-fluorobenzyl)-2-(4-fluorophenyl)-N-piperidin-4-yl- acetamide was prepared in the same way as 103NLS56.
[0647] R/ = 0.26 (MeOH/CH2Cl2, 5:95). LCMS m/z 459 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.28-6.91 (m, 8H5 Ar-H)5 4.63-4.52 (m, 0.6H5 pip-H), 4.51 & 4.46 (2s5 2H, benzyl-H), 3.92-3.88 (m, 2H, dioxane-H), 3.82-3.69 (m, 3.2 H5 dioxane-H, benzyl-H, pip-H), 3.54 (s, 1.2H, benzyl-H), 2.99-2.90 (m, 2H5 pip-H). 2.62-2.51 (m, IH, dioxane-H), 2.39-2.28 (m, 1.2H, pip-H), 2.18-2.10 (m, 0.8H, pip-H), 1.72-1.50 (m, 3.2H, pip-H), 1.42-1.31 (m, 6.8H, piρ-H5 dioxane-CH3). HPLC tκ = 7.9 min.
Example 191 - N-Cl-r2-(1.3-Dioxan-4-yl)ethyllpiperidin-4-vn-N-f4-fluorobenzvn-2-r4- fluorophenyTjacetamide, tartrate (130AF41-171). [0648] The starting material W-(4-fluorobenzyl)-2-(4-fluorophenyl)-iV- piperidin-4-yl-acetamide was prepared in the same way as 103NLS56.
[0649] Potassium carbonate (64 mg, 0.46 mmol) was added to a solution of N- (4-fluorobenzyl)-2-(4-fluorophenyl)-N-piperidin-4-yl-acetamide (79.4 mg, 0.23 mmol) in dry ΛyV-dimethylformamide (3 rtiL). To this suspension a solution of 4-[2- (tosyloxy)ethyl]-l ,3-dioxane 128NLS46B (99 mg, 0.35 mmol) in dry N1N- dimethylformamide (1 mL) was added dropwise at rt. The reaction mixture was stirred overnight at 600C and it was partitioned between dichloromethane and water. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with stepwise gradient of 2-5% methanol in dichloromethane, afforded the desired product (71 mg, 67%). The product was converted to its tartrate form as described above.
[06501 R/ = 0.41 (MeOH/CH2Cl2, 6:96). LCMS m/z 459 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.28-6.90 (m, 8H, Ar-H)5 4.99 (m, IH, dioxane-H), 4.77-4.64 (m, 1.6H, pip-H, dioxane-H), 4.52 (s, 2H, benzyl-H), 3.80-3.56 (m, 4.4H, dioxane-H, benzyl-H, pip-H), 3.21-3.08 (m, 1.2H, pip-H), 2.96-2.88 (m, 0.8H, pip-H), 2.75-2.56 (m, 1.2H, NCH2), 2.52-2.24 (m, 2H, pip-H, NCH2), 2.04-1.30 (m, 9.8H5 pip-H, NCH2CH2, dioxane-H). HPLC /R = 6.4 min.
Example 192 - N-ll-f2-ri.3-Dioxan-4-vnethyllpiτ)eridin-4-yl>-N-f4-fluorobenzylV2-r4- trifluoromethoxyphenvDacetamide. tartrate (130AF80-186').
[0651] Triethylamine (125 μL, 0.89 mmol) was added to a solution of N-{ \- [2-(l,3-dioxan-4-yl)ethyl]piperidin-4-yl}-iV-(4-iluorobenzyl)amine 128NLS52 (96 mg, 0.30 mmol) in dry dichloromethane (5 mL) at rt. The solution was cooled to — 100C and a solution of (4-trifluoromethoxyphenyl)acetyl chloride (71 mg, 0.30 mmol) in dry dichloromethane (1 mL) was added dropwise. The reaction mixture was stirred overnight at rt. The solvent was removed by evaporation under reduced pressure. The residue was suspended in tetrahydrofuran and filtered. The filtrate was evaporated to dryness and the residue was purified by silica gel column chromatography, eluting with 4% methanol in dichloromethane, to give the desired compound (46 mg, 30%). The compound was converted to its tartrate form as described above. [06521 R/ = 0.33 (MeOH/CH2Cl2, 6:94). LCMS m/z 459 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.34-6.91 (m, 8H5 Ar-H), 5.01 (d, IH5 J = 6.0, dioxane-H)5 4.66-4.54 (m, 1.6H, pip-H, dioxane-H)5 4.52 & 4.49 (2s, 2H, benzyl-H), 4.09-4.05 (ra, IH, dioxane-H), 3.83 (s, 0.8H5 benzyl-H)5 3.72-3.56 (m, 3.6 H, dioxane-H, benzyl-H5 pip- H)5 2.94-2.86 (m, 2H, pip-H), 2.50-2.32 (m, 2H5 NCH2), 2.11-2.00 (m, 1.2H5 pip-H), 1.90-1.52 (m, 8.8H5 pip-H, NCH2CH2, dioxane-H). HPLC tR= 7.6 min.
Example 193 - N-π-r2-d,3-Dioxan-4-vnethyllpiperidin-4-vU-N-f4-fluorobenzvn-2-r4- propoxyphenvPacetamide, tartrate (130AF71-184).
[0653] Triethylamine (163 μL, 1.17 mmol) was added to a solution of N- {I - [2-(l53-dioxaπ-4-yl)ethyl]piperidin-4-yl}-N-(4-fluoroben2yl)amine 128NLS52 (126 mg, 0.39 mmol) in dry dichloromethane (5 mL) at rt. The solution was cooled to -150C and a solution of (4-propoxyphenyl)acetyl chloride (92 mg, 0.43 mmol) in dry dichloromethane (2 mL) was added dropwise. The reaction mixture was stirred for 2 hours at rt. The solvent was removed by evaporation under reduced pressure. The residue was suspended in tetrahydrofuran and filtered. The filtrate was evaporated to dryness and the residue was purified by silica gel column chromatography, eluting with a stepwise gradient of 0- 4% methanol in dichloromethane, to give the desired compound (66 mg, 34%). The product was converted to its tartrate form as described above.
[0654] R7 = 0.16 (MeOH/CH2Cl25 4:96). LCMS m/z 499 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.21-6.78 (m5 8H, Ar-H)5 5.00 (m, IH, dioxane-H), 4.66-4.54 (m, 1.6H, pip-H, dioxane-H), 4.50 & 4.44 (2s, 2H, benzyl-H), 4.10-4.03 (m, IH, dioxane- H), 3.92-3.87 (m, 2H5 OCH20Pr), 3.78-3.50 (m, 4.4 H5 dioxane-H, benzyl-H, pip-H), 2.92- 2.82 (m, 2H, pip-H), 2.50-2.29 (m, 2H, NCH2), 2.09-1.98 (m, 1.2H, pip-H), 1.88-1.27 (m, 10.8H, ρip-H, NCH2CH2, dioxane-H, CH2Opr), 1.05-099 (m, 3H, CH3OPr). HPLC tR = 7.6 min.
Example 194 - N-(4-Fluorobenzyl)-2-r4-isobutoxyphenyl')-N-ri-('tetrahvdropyran-4- yl)piperidin-4-yl1acetamide. tartrate (130AF33-166).
[0655J A solution of tetrahydro-4H-pyran-4-one (43 mg, 0.42 mmol) in methanol (1 mL) was added to a solution of 103NLS56 (57 mg, 0.14 mmol) in methanol (2 mL). After addition of acetic acid (100 μL) the reaction mixture was stirred for 15 minutes in a sealed flask under microwave irradiation at 1000C. Afterwards sodium cyanoborohydride (26 mg, 0.42 mmol) was added to the mixture and stirring was continued for additional 60 min under microwave irradiation at 800C. The mixture was passed over an acidic ion-exchange SPE cartridge. Further purification of the product by silica gel column chromatography, eluting with a stepwise gradient of 2-5% methanol in dichloromethane, afforded the desired compound (19.2 mg, 28%). The compound was converted to its tartrate form as described above.
[0656] R/ = 0.18 (MeOH/CH2Cl2, 5:95). LCMS m/z 483 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.21-6.80 (m, 8H, Ar-H), 4.64-4.56 (m, 0.6H, pip-H), 4.51& 4.45 (2s, 2H, benzyl-H), 4.02-3.96 (m, 2H5 THP-H), 3.77-3.68 (m, 3.2H, benzyl-H, CH2OiBu, pip-H), 3.51 (s, 1.2H, benzyl-H), 3.30 (t, 2H, J = 12.0, THP-H), 2.98-2.88 (m, 2H, pip-H), 2.46-2.34 (m, IH, THP-H)5 2.28-2.19 (m, 1.2H, pip-H), 2.10-1.99 (m, 1.8H, CHoiBu, pip-H), 1.73-1.47 (m, 7.2H, pip-H, THP-H), 1.39-1.33 (m, 0.8H, pip-H), 1.01 (m, 6H, CH3OiBu). HPLC tR= 8.0 min.
Example 195 - N-f4-Fluorobenzyl)-2-('4-isobutoxyphenvπ-N-fl-rtetrahvdropyran-4- ylmethvπpiperidin-4-yl]acetamide, tartrate (130AF82-187)
[0657] The title compound was synthesized from N-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)-iV-piperidin-4-yl-acetamidel03NLS56 (110 mg, 0.27 mmol) and tetrahydro-2H-pyran-4-yl carbaldehyde (63 mg, 0.55 mmol) using the same method as for preparation of 130AF33-166. Yield: 18 mg, 13%.
[0658] Rf = 0.30 (MeOH/CH2Cl2, 5:95). LCMS m/z 497 [M+H]+. HPLC /R = 8.4 min.
Example 196 - N-(4-FluorobenzylV2-r4-isobutoxyphenyl)-N-ri-r2-rtetrahydropyran-4- vDethyllpiperidin-4-vHacetarnide, tartrate (130AF83-188*)
[0659] The title compound was synthesized from Λ/-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)-N-piperidm-4-yl-acetamidel03NLS56 (110 mg, 0.27 mmol) and tetrahydro-2H-pyran-4-yl acetalaldehyde (70.5 mg, 0.55 mmol) using the same method as for preparation of 130AF33-166. Yield: 40 mg, 29%.
[0660] R7 = 0.30 (MeOH/CH2Cl2, 5:95). LCMS m/z 511 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.21-6.80 (m, 8H, Ar-H), 4.65-4.55 (m, 0.6H, pip-H), 4.51& 4.44 (2s, 2H, benzyl-H), 3.95-3.89 (m, 2H5 THP-H), 3.78-3.66 (m, 3.2H, benzyl-H, CH2OiBu, pip-H), 3.51 (s, 1.2H5 benzyl-H), 3.34 (t, 2H, J = 12.0, THP-H), 2.92-2.82 (m5 2H, pip-H), 2.34-2.26 (m, 2H, NCH2CH2), 2.11-1.96 (m, 2.2H, pip-H, CHOIBU), 1.84-1.20 (m, 11.8H, pip-H, THP-H, CH2CH2N), 1.02 (m, 6H5 CH3OiBu). HPLC fe = 8.2 min.
Example 197 - N-(4-'Fluorobenzviy2-f4-fluorophenylVN-[l-ftetrahydropwan-4- vl)piperidin-4-vπacetamide. tartrate ( 130AF37-169V
[0661] The desired compound was synthesized from tetrahydro-4H-pyran-4- one and N-(4-fluorobenzyl)-2-(4-fluorophenyl)-N-piperidin-4-yl-acetamide using the same method as for preparation of 130AF33-166. The starting material iV-(4- fluorobenzyl)-2-(4-fluorophenyl)-N-piperidin-4-yl-acetamide was prepared in the same way as 103NLS56.
[0662] R/ = 0.29 (MeOH7CH2Cl2, 5:95). LCMS m/z 429 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.29-6.91 (m, 8H, Ar-H), 4.64-4.55 (m, 0.6H, pip-H), 4.52 & 4.48 (2s, 2H5 benzyl-H), 4.02-3.95 (m, 2H, THP-H)5 3.80 (s, 0.8H, benzyl-H), 3.75-3.64 (m, 0.4H, pip-H), 3.54 (s, 1.2H5 benzyl-H), 3.34 (t, 2H, J= 12.0, THP-H), 2.99-2.90 (m, 2H, pip-H), 2.48-2.36 (m5 IH, THP-H), 2.26-2.20 (m, 1.2H, pip-H), 2.08-2.00 (m, 0.8H5 pip-H), 1.76-1.47 (m5 7.2H, pip-H5 THP-H)5 1.41-1.34 (m5 0.8H, pip-H). HPLC fR = 5.6 min.
Example 198 - N-ri-f(SV3.5-Dihydroxypentvnpiperidine-4-yl1-N-f4-fluorobenzylV2-(4- isobutoxyphenvDacetamide, tartrate (130AF65-182).
[0663] The compound (i?)-5-[(4-methylbenzenesulfonyl)oxy]pentane-l,3-diol was synthesized according to Moune et άl (J.Org.Chem., 1997, 62, 3332-3339). Potassium carbonate (83 mg, 0.60 mmol) was added to a solution of 103NLS56 (94 mg, 0.24 mmol) in dry JV,N-dimethylformamide (3 mL). To this suspension a solution of (R)- 5-[(4-methylbenzenesulfonyl)oxy]pentane-l,3-diol (82 mg, 0.28 mmol) in dry NJV- dimethylformamide (1 mL) was added, followed by addition of sodium iodide (43 mg, 0.29 mmol). The reaction mixture was stirred overnight at 600C. It was allowed to cool to rt, filtered and evaporated to dryness. The residue was partitioned between dichloromethane and 2M aq sodium hydroxide. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with a stepwise gradient of 6-10% methanol in dichloromethane, afforded the desired compound (35 mg, 29%), which was converted to its tartrate form as described above.
[0664] Rf= 0.48 (MeOH/CH2Cl2> 10:90). LCMS m/z 501 [M+H]+. HPLC fo = 7.4 min.
Example 199 - N-π-r2-f(4SV13-Dioxane-4-vnethvnpiperidine-4-vU-N-f4- fluorobenzyr)-2-(4-isobutoxyphenyDacetamide, tartrate (130AF67-183).
[0665] Paraformaldehyde (9 mg, 0.28 mmol) and hydrochloric acid (4M in 1.4-dioxane, 0.5 mL) were added to a solution of tartaric acid salt of 130AF65-182 (37 mg, 0.056 mmol) in 1.4-dioxane. The reaction mixture was stirred for 2 hours in a sealed flask under microwave irradiation at 1200C and partitioned between dichloromethane and sodium bicarbonate. The organic layer was washed with brine, dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by acidic ion- exchange SPE cartridge afforded the desired compound (9.0 mg, 31%), which was converted to its tartrate form as described above. The enantiomeric excess (ee) was determined to be 94% using chiral HPLC analysis (Chiralpak AD column, 4.6 x 250 mm; heptane/I-PrOH 50:50, 0.3% DEA; 0.5 mL/min; tR 20.5 min).
[0666] Rf = 0.41 (MeOH/CH2Cl2, 8:92). LCMS m/z 513 [M-HH]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.21-6.80 (m, 8H, Ar-H), 5.00 (m, IH5 dioxane-H), 4.68-4.54 (m, 1.6H5 pip-H, dioxane-H), 4.51 & 4.45 (2s, 2H, benzyl-H), 4.06 (m, IH, dioxane-H), 3.77-3.48 (m, 6.4H, dioxane-H, benzyl-H, CH201Bu, piρ-H)5 2.98-2.79 (m, 2H, pip-H), 2.50-2.58 (m, 2H, NCH2), 2.14-1.99 (m, 2.2H, CHOlBu, pip-H), 1.90-1.25 (m, 8.8H, pip-H, NCH2CH2, dioxane-Η)5 1.02 (m, 6H, CH3O.BU). HPLC /R= 8.7 min.
Example 200 - N-π-f2-(1.3-Dioxan-2-yl)ethvnpiperidin-4-vU-N-(4-fluorobenzyl') amine fl l8AF52-95\
[0667] Sodium carbonate (17.4 g5 125.9 mmol) was added to a solution of 4- piperidone monohydrate hydrochloride (6.45 g, 42.0 mmol) in acetonitrile (200 mL). After 30 minutes stirring at rt a solution of 2-(2-bromoethyl)-l,3-dioxane (8.45 g, 43.3 mmol) in acetonitrile (50 mL) was added dropwise to the reaction mixture and stirring was continued overnight at rt and at reflux for an additional 2 hours. The solvent was removed by evaporation under reduced pressure and the residue was partitioned between water and dichloromethane. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with 7% methanol in dichloromethane, afforded l-[2-(l,3- dioxan-2-yl)ethyl]piperidin-4-one (6.19 g, 69%).
[0668] A solution of l-[2-(l,3-dioxan-2-yl)ethyl]piperidin-4-one (6.19 g, 29 mmol) in methanol (80 mL) was added dropwise to a solution of 4-fluorobenzylamine (3.9 mL, 34 mmol) in methanol (100 mL) under argon atmosphere at rt. After 30 minutes stirring at rt the reaction mixture was acidified (pH = 5) with acetic acid and cooled to 00C. Sodium cyanoborohydride (2.15 g, 40 mmol) was added slowly to the cold mixture and stirring was continued at rt overnight. The reaction mixture was basified with 2M NaOH and concentrated in vacuo. The residue was partitioned between ethyl acetate and water. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. The residue was dissolved in abs. ethanol (57 mL). A solution of maleic acid (3.31 g, 28.5 mmol) in abs. ethanol (60 mL) was added to this solution resulting in precipitate formation. The precipitate was collected by filtration and converted to the free base by a basic extraction. Yield: 8.5 g, 91%.
[0669] R/ = 0.29 (MeOH/CH2Cl2, 7:93). LCMS m/z 323 [M+H]+.1H NMR (CDCl3) δ 7.25 (m, 2H5 Ar-H), 6.95 (m, 2H5 Ar-H)3 4.54 (t, IH, J= 5.6, dioxane-H), 4.07- 4.02 (m5 2H, dioxane-H), 3.73-3.67 (m, 4H, dioxane-H, benzyl-H), 2.85-2.79 (m, 2H, pip- H)5 2.49-2.37 (m, 3H, NCH2, ρiρ-H), 2.05-1.72 (m5 7H, piρ-H, NCH2CH2, dioxane-H), 1.44-1.25 (m, 4H, dioxane-H, pip-H, NH). HPLC /R= 1.4 min.
Example 201 - 2-(4-BBnZvIoXVPhBnVlVN- H- [2-Cl.3-dioxan-2-vnethylipiperidin-4-vU -N- f4-fluorobenzyr)acetamide. tartrate (1 18AF66-102).
[0670] A solution of triethylamine (0.89 mL, 6.38 mmol) and 1 18AF52-95 (0.80 g, 2.48 mmol) in dry THF (10 mL) was cooled to 00C. A solution of 4- benzyloxyphenylacetyl chloride (0.72 g, 2.76 mmol) was added dropwise to the cold reaction mixture and stirring was continued at rt for 2 h. The reaction mixture was filtered and the filtrate evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with a stepwise gradient of 0-6% methanol in dichloromethane afforded the desired compound (0.53 g, 39%), which was converted to its tartrate form as described above. [0671] R/ = 0.27 (MeOH/CH2Cl2, 7:93). LCMS m/z 547 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.46-6.86 (m, 13H5 Ar-H)5 5.08-5-02 (m, 2H5 PhCH2O)5 4.64- 4.42 (m, 3.6H5 pip-H, benzyl-H, dioxane-H), 4.11-4.02 (m, 2H5 dioxane-H)5 3.79-3.67 (m, 3.2H5 dioxane-H, benzyl-H, pip-H), 3.50 (s, 1.2H, benzyl-H), 2.94-2.80 (m, 2H, ρip-H), 2.46-2.34 (m, 2H5 NCH2), 2.12-1.98 (m, 2.2H5 dioxane-H, pip-H), 1.87-1.50 (m5 6H, pip- H, NCH2CH2), 1.36-1.24 (m, 1.8H, pip-H, dioxane-H). HPLC rR= 8.9 min.
Example 202 - N-(l-r2-π.3-Dioxan-2-yl)ethyllpiperidin-4-γU-N-(4-fluorobenzyl)-2-(4- hvdroxyphenvπ-acetamide, tartrate Cl 18AF67-1031
[0672] The desired compound was afforded by hydrogenation of 118AF66- 102 (0.50 g, 0.92 mmol) in absolute ethanol (200 mL) using palladium on carbon as a catalyst. The product was purified by column chromatography on silica gel eluting with a stepwise gradient of 3-6 % methanol in dichloromethane. The desired compound (0.22 g, 53 %) was converted to its tartrate form as described above.
[0673] R/ = 0.30 (MeOH/CH2Cl2, 6:94). LCMS m/z 457 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.13-6.86 (m, 6H5 Ar-H), 6.72-6.64 (m, 2H, Ar-H)5 4.66-4.57 (m5 0.6H, pip-H), 4.54 (m, IH, dioxane-H), 4.48 & 4.37 (2s, 2H5 benzyl-H), 4.08-4.01 (m, 2H, dioxane-H), 3.80-3.66 (m, 3.2H, dioxane-H, benzyl-H, pip-H), 3.47 (m, 1.2H, benzyl- H), 2.94-2.82 (m, 2H, pip-H), 2.47-2.39 (m5 2H, NCH2), 2.10-1.97 (m, 2.2H, dioxane-H, pip-H), 1.88-1.53 (m5 6H, pip-H, NCH2CH2), 1.34-1.25 (m, 1.8Η, pip-H, dioxane-H). HPLC ZR = 3.0 min.
Example 203 - N-n-r2-fl.3-Dioxan-2-vnethvnpiperidin-4-vn-N-(4-fluorobenzvn-2-(4- methoxyphenylVacetamide, tartrate (118AF60-96\
[0674] A solution of triethylamine (0.57 mL, 4.09 mmol) and 1 18AF52-95 (328 mg, 1.02 mmol) in dry THF (5 mL) was cooled to 00C. A solution of 4- methoxyphenylacetyl chloride (376 mg5 2.04 mmol) was added dropwise to the cold reaction mixture and stirring was continued for 20 h at rt. The reaction mixture was partitioned between 2M NaOH and water. The organic layer was dried over sodium sulphate, filtered and evaporated to dryness. The residue was purified by silica gel column chromatography, eluting with a stepwise gradient of 0-6 % methanol in dichloromethane. Final purification of the product by acidic ion-exchange SPE cartridge afforded the desired compound (153 mg, 33 %), which was converted to its tartrate form as described above.
[0675J R/ = 0.40 (MeOH/CH2Cl2, 4:96). LCMS m/z Al\ [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.24-6.79 (m, 8H, Ar-H), 4.63-4.54 (m, 0.6H3 ρip-H), 4.52 (t, IH, J= 5.2, dioxane-H), 4.49 & 4.44 (2s, 2H, benzyl-H), 4.09-4.01 (m, 2H5 dioxane-H), 3.79-3.68 (m, 6.2H5 dioxane-H, benzyl-H, piρ-H, OCH3), 3.50 (m, 1.2H, benzyl-H), 2.91- 2.80 (m, 2H5 ρiρ-H), 2.43-2.36 (m, 2H, NCH2), 2.10-1.98 (m, 2.2H, dioxane-H, pip-H), 1.86-1.51 (m, 6H, pip-H, NCH2CH2), 1.34-1.26 (m, 1.8H, pip-H, dioxane-H). HPLC /R = 7.0 min.
Example 204 - N-(l-r2-π,3-Dioxan-2-yl)ethyl1ρiperidin-4-vU-N-C4-fluorobenzyl')-2-(4- isopropylphenylVacetamide, tartrate (118AF63-100).
[0676] The desired compound was synthesized from 118AF52-95 (400 mg, 1.24 mmol) and 4-isopropylphenylacetyl chloride (340 mg, 1.73 mmol) using the same method as for preparation of 118AF66-102. Further purification by acidic ion-exchange SPE cartridge was performed. Yield: 273 mg, 46 %.
[0677] R7 - 0.34 (MeOH/CH2Cl2, 7:93). LCMS m/z 483 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.22-6.89 (m, 8H, Ax-H), 4.64-4.43 (m, 3.6H5 pip-H, dioxane- H, benzyl-H), 4.09-4.02 (m, 2H, dioxane-H), 3.79 (s, 0.8H, benzyl-H), 3.76-3.66 (m, 2.4H5 dioxane-H, pip-H), 3.54 (m, 1.2H, benzyl-H), 2.92-2.79 (m, 3H, pip-H, C7/(CH3)2), 2.41-2.35 (m, 2H, NCH2), 2.12-1.98 (m, 2.2H, dioxane-H, pip-H), 1.85-1.49 (m, 6H, pip- H, NCH2CH2), 1.34-1.19 (m, 7.8H, pip-H, dioxane-H, CH(CH^). ΗPLC fR= 8.6 min.
Example 205 - N-π-r2-π.3-Dioxan-2-vnethvnpiperidin-4-yll-N-f4-fluorobenzyl)-2-r4- trifluoromethoxy-phenvDacetarnide. tartrate Cl 18AF58-98").
[0678] The desired compound was synthesized from 118AF52-95 (328 mg, 1.02 mmol) and 4-trifluoromethoxyphenylacetyl chloride (345 mg, 1.44 mmol) using the same method as for preparation of 118AF66-102. Yield: 267 mg, 49 %.
[0679] R/ = 0.31 (MeOΗ/CΗ2Cl2, 4:96). LCMS m/z 525 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.30-6.90 (m, 8H, Ar-H), 4.63-4.48 (m, 3.6H, pip-H, dioxane- H, benzyl-H), 4.05 (m, 2H, dioxane-H), 3.82 (s, 0.8H5 benzyl-H), 3.76-3.62 (m, 2.4H, dioxane-H, pip-H), 3.55 (m, 1.2H, benzyl-H), 2.92-2.84 (m, 2H, pip-H), 2.43-2.36 (m, 2H, NCH2), 2.10-1.96 (m, 2.2H, dioxane-H, pip-H), 1.88-1.79 (m, 0.8H, pip-H), 1.76- 1.52 (m, 5.2H, pip-H, NCH2CH?), 1.38-1.26 (m, L8Η, pip-H, dioxane-H). HPLC tø= 8.4 min.
Example 206 - N-d-r∑-d.S-Dioxan^-vnethvnpiperidin-^vU-N-^-fluorobenzylVΣ^- ethoxyphenvD-acetamide. oxalate (118AF68-104).
[0680] The desired compound was synthesized from 118AF52-95 (400 mg, 1.24 mmol) and 4-ethoxyphenylacetyl chloride (300 mg, 1.51 mmol) using the same method as for preparation of 118AF66-102. Further purification by acidic ion-exchange SPE cartridge was performed. Yield: 0.15 g, 25 %.
[0681] R7 = 0.26 (MeOH/CH2Cl2, 6:94). LCMS m/z 485 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.20-6.79 (m, 8H, Ar-H), 4.64-4.54 (m, 0.6H, pip-H), 4.52 (t, IH, J = 5.2, dioxane-H), 4.49 & 4.43 (2s, 2H, benzyl-H), 4.07-3.97 (m, 4H5 dioxane-H, OCH2), 3.76-3.66 (m, 3.2H, dioxane-H, pip-H, benzyl-H), 3.49 (s, 1.2H, benzyl-H), 2.91- 2.80 (m, 2H, pip-H), 2.42-2.32 (m, 2H, NCH2), 2.10-1.97 (m, 2.2H, dioxane-H, pip-H), 1.86-1.48 (m, 6H5 pip-H, NCH2CH2), 1.42-1.36 (m, 3H, CH3), 1.34-1.24 (m, 1.8H, ρip-H5 dioxane-H). HPLC fR= 7.6 min.
Example 207 - N-n-r2-ri.3-Dioxan-2-vnethyliDiperidin-4-vn-N-C4-fluoroben2ylV2-(4- isopropoxyphenyP-acetamide, oxalate (118AF73-107).
[0682] The desired compound was synthesized from 118AF52-95 (400 mg, 1.24 mmol) and 4-isopropoxyphenylacetyl chloride (340 mg, 1.60 mmol) using the same method as for preparation of 118AF66-102. Further purification by acidic ion-exchange SPE cartridge was performed. Yield: 91 mg, 15 %.
[0683] R/ = 0.58 (MeOHZCH2Cl25 8:92). LCMS m/z 499 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.19-6.78 (m, 8H, Ar-H), 4.64-4.42 (m, 4.6H, pip-H, dioxane- H5 benzyl-H, CHoiPr), 4.07 (m, 2H, dioxane-H), 3.76-3.68 (m, 3.2H, dioxane-H, pip-H, benzyl-H)5 3.49 (s, 1.2H5 benzyl-H), 2.91-2.80 (m, 2H, pip-H), 2.42-2.35 (m, 2H, NCH2), 2.10-1.99 (m, 2.2H, dioxane-H, pip-H), 1.85-1.51 (m, 6H, pip-H, NCH2CH2), 1.31 (m, 7.8Η, OCH(CHj)2, pip-Η, dioxane-Η). ΗPLC rR = 8.1 min.
Example 208 - N-π-r2-ri.3-Dioxan-2-vnethyllDiperidin-4-vn-N-r4-fluorobenzylV2- phenylacetamide. oxalate (\ 18AF77-109V [0684] The desired compound was synthesized from 118AF52-95 (300 mg, 0.93 mmol) and phenylacetyl chloride (197 mg, 1.27 mmol) using the same method as for preparation of 118AF66-102. Further purification by acidic ion-exchange SPE cartridge was performed. Yield: 68 mg, 17 %.
[0685] R/ = 0.28 (MeOH/CH2Cl2, 5:95). LCMS m/z 441 [MH-H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.33-6.89 (m, 9H, Ar-H), 4.65-4.44 (m, 3.6H, piρ-H, dioxane- H, benzyl-H), 4.09-4.03 (m, 2H, dioxane-H), 3.84 (s, 0.8H, benzyl-H), 3.76-3.67 (m, 2.4H, dioxane-H, pip-H), 3.57 (s, 1.2H, benzyl-H), 2.92-2.79 (m, 2H, pip-H), 2.44-2.34 (m, 2H, NCH2), 2.10-1.98 (m, 2.2H, dioxane-H, pip-H), 1.86-1.51 (m, 6H, pip-H, NCH2CH2), 1.34-1.23 (m, 1.8Η, pip-H, dioxane-H). HPLC JR = 6.1 min.
Example 209 - N-{l-r2-(1.3-Dioxan-2-vnethyllpiperidin-4-vU-N-(4-fluorobenzylV2-r4- re-fluoroethoxyVphenvπacetamide. oxalate (118AF85-113).
[0686] The desired compound was synthesized from 118AF52-95 (360 mg, 1.11 mmol) and 4-(2-fluoroethoxy)phenylacetyl chloride (282 mg, 1.30 mmol) using the same method as for preparation of 118AF66-102. Further purification by acidic ion- exchange SPE cartridge was performed. Yield: 84 mg, 15 %.
[0687] R/ = 0.36 (MeOH/CH2Cl2, 5:95). LCMS m/z 503 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.27-6.84 (m, 8H, Ar-H), 4.80 (m, IH, OCH2CH2F), 4.68 (m, 1Η, OCH2CH2F), 4.65-4.45 (m, 3.6H, pip-H, dioxane-H, benzyl-H), 4.22 (m, IH, OCH2CH2F), 4.16 (m, IH, OCH2CH2F), 4.10-4.03 (m, 2H, dioxane-H), 3.79-3.68 (m, 3.2H, dioxane-H, pip-H, benzyl-H), 3.51 (s, 1.2H, benzyl-H), 2.92-2.82 (m, 2H, pip-H), 2.44-2.36 (m, 2H, NCH2), 2.12-1.99 (m, 2.2H, dioxane-H, pip-H), 1.88-1.51 (m, 6H, pip- H, NCH2CH2), 1.35-1.26 (m, 1.8H, pip-H, dioxane-H). HPLC ^R = 7.0 min.
Example 210 - N-(l-r2-r5.5-Dimethyl-1.3dioxan-2-vnethyllpiperidin-4-vU-N-(4- fluorobenzyl)-2-(4-isobutoxyphenyl)acetamide, oxalate d 18AF27-83).
[0688] The desired compound was synthesized from _V-{l-[2-(l,3-dioxan-2~ yl)ethyl]piperidin-4-yl } -N-(4-fluorobenzyl)-2-(4-isobutoxyphenyl)acetamide 103NLS63F (22 mg, 0.042 mmol) and 2,2-dimethyl-l53-propandiol (33 mg, 0.38 mmol) using the same method as for preparation of 130AF 12- 148. Purification of the product by reversed phase HPLC (Q g) afforded the title compound (2.8 mg, 12 %). LCMS m/z 541 [M+H]+. HPLC fR= 9.9 min. Example 211 - N-r4-FluorobenzylV2-r4-isobutoxyDhenvn-N-n-r2-rfRV4-methyl-1.3- dioxan-2-vnethyl1-piperidin-4-vUacetamide. oxalate d l8AF29-84>.
[0689] The desired compound was synthesized from iV-{l-[2-(l,3-dioxan-2- yl)ethyl]piperidin-4-yl } -N-(4-fluorobenzyl)-2-(4-isobutoxyphenyl)acetamide 103NLS63F (38 mg, 0.074 mmol) and (i?)-(-)-l,3-butandiol (33 mg, 0.38 mmol) using the same method as for preparation of 130AF12-148. Purification of the product by reversed phase HPLC (Ci8) afforded the title compound (11.6 mg, 28 %). LCMS m/z 527 [M+H]+. HPLC /R = 8.7 min.
Example 212 - N-^-Fluorobenzvn^-^-isobutoxyphenvD-N-π-r∑-ffS^^-methyl-l.S- dioxolan-2-yDethyl"|-piperidin-4-yl}acetamide, oxalate ( 118AF31-85*).
[0690] The desired compound was synthesized from iV-{l-[2-(l,3-dioxan-2- yl)ethyl]piperidin-4-yl}-N-(4-fluoroben2yl)-2-(4-isobutoxyphenyl)acetamide 103NLS63F (40 mg, 0.078 mmol) and (S)-(+)-propylene glycol (30 mg, 0.39 mmol) using the same method as for preparation of 130AF12-148. Purification of the product by reversed phase HPLC (Ci8) afforded the title compound (21 mg, 53 %). LCMS m/z 513 [M+H]+. HPLC /R = 9.9 min.
Example 213 - N-ll-r∑-^.ό-Dimethyl-l.S-dioxan^-vπethylipiperidin^-vU-N-^- fluorobenzvD-2-f4-isobutoxyphenyl)acetamide, oxalate (\ 18AF37-88').
[0691] The desired compound was synthesized from N-{l-[2-(l,3-dioxan-2- yl)ethyl]piperidin-4-yl}-iV-(4-fluorobenzyl)-2-(4-isobutoxyphenyl)acetamide 103NLS63F (40 mg, 0.078 mmol) and 2,4-pentandiol (41 mg, 0.39 mmol) using the same method as for preparation of 13 OAF 12- 148. Purification of the product by reversed phase HPLC (Ci8) afforded the title compound (9 mg, 21 %). LCMS m/z 541 [M+H]+. HPLC fR= 10.5 min.
Example 214 - N-f4-Fluoroben2yl')-N-fl-r2-frSV4-methyl-1.3-dioxolan-2-vDethyll piperidin-4-yl)-2-(4-trifluoromemoxyphenvDacetamide. oxalate Cl 18AF87-114V
[0692] The desired compound was synthesized from jV-{l-[2-(l,3-dioxan-2- yl)ethyl]piperidin-4-yl}-7V-(4-fluorobenzyl)-2-(4-trifluoromethoxyphenyl)acetamide 118AF58-98 (70 mg, 0.13 mmol) and (5)-(+)-propylene glycol (53 mg, 0.69 mmol) using the same method as for preparation of 130AF 12- 148. Purification of the product by silica gel column chromatography, eluting with a stepwise gradient of 0-4% methanol in dichloromethane, afforded the title compound (31 mg, 46%). R/ = 0.17 (MeOH/CH2Cl2 4:96). LCMS m/z 525 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.34-6.91 (m, 8H, Ar-H), 5.03 & 4.92 (2t, IH5 J = 4.8, dioxolane-H), 4.66-4.56 (m, 0.6H, pip-H), 4.52 & 4.49 (2s, 2H, benzyl-H), 4.22-4.07 (m, 1.4H, dioxolane-H), 3.95-3.89 (m, 0.6H5 dioxolane-H), 3.84 (s, 0.8H5 benzyl-H)5 3.74-3.64 (m, 0.4H, pip-H), 3.57 (s, 1.2H, benzyl- H), 3.40-3.33 (m, IH, dioxolane-H), 2.78-2.86 (m, 2H, pip-H), 2.49-2.38 (m5 2H5 NCH2), 2.10-2.01 (m, 1.2H5 pip-H), 1.70-1.53 (ra, 6H, pip-H5 NCH2CH2), 1.40-1.22 (m, 3.8H, pip-H, CH3). HPLC tR= 8.7 min.
Example 215 - N-f4-Fluorobenzviy2-f4-isopropylphenylVN-π-j7-ffSV4-methyl-1.3- dioxolan-2-yl')ethyl')-piperidin-4-yl)acetamide. oxalate (118AF91-1 17).
[0693] The desired compound was synthesized from N-{l-[2-(l,3-dioxan-2- yl)ethyl]piperidin-4-yl}-iV-(4-fluorobenzyl)-2-(4-isopropylphenyl)acetamide 118AF63- 100 (150 mg, 0.31 mmol) and (iS>(+)-propylene glycol (95 mg, 1.24 mmol) using the same method as for preparation of 130AF 12- 148. Purification by silica gel column chromatography, eluting with a stepwise gradient of 0-4 % methanol in dichloromethane, afforded the title compound (51.2 mg, 34%).
10694] R/ = 0.19 (MeOH/CH2Cl2, 4:96). LCMS m/z 483 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.24-6.90 (m, 8H, Ar-H), 5.03 & 4.92 (2t, IH, J = 4.8, dioxolane-H), 4.67-4.55 (m, 0.6H, pip-H), 4.51 & 4.47 (2s, 2H, benzyl-H), 4.21-4.07 (m, 1.4H, dioxolane-H), 3.94-3.89 (m, 0.6H, dioxolane-H), 4.81-3.50 (m, 1.2H, benzyl-H, pip-H), 3.55 (s, 1.2H, benzyl-H), 3.40-3.33 (m, IH, dioxolane-H), 2.94-2.83 (m, 3H, pip- H, CH(CHj)2), 2.47-2.38 (m, 2H, NCH2), 2.09-2.01 (rn, L2, pip-H), 1.86-1.52 (m, 6H, pip-H, NCH2CH2), 1.31-1.19 (m, 9.8H, pip-H, CH3, CH(CH3)2). HPLC tR= 8.6 min.
Example 216 - N-(4-Fluorober^lVN-π-r2-((RV4-methyl-13-dioxan-2-vnethyπ piperidin-4-yl}-2-(4-trifluoromethoxyphenyl)acetamide, oxalate (I 18AF75-108).
[0695] The desired compound was synthesized from iV-{l-[2-(l,3-dioxan-2- yl)ethyl]piperidin-4-yl}-N-(4-fluorobenzyl)-2-(4-trifluoromethoxyphenyl)acetamide 118AF58-98 (70 mg, 0.13 mmol) and (K)-(-)-l,3-butandiol (60 mg, 0.66 mmol) using the same method as for preparation of 130AF12-148. Purification of the product by silica gel column chromatography, eluting with a stepwise gradient of 0-4 % methanol in dichloromethane, afforded the title compound (28 mg, 40%).
[0696] R/ = 0.24 (MeOH/CH2Cl2, 5:95). LCMS m/z 539 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.33-6.91 (m, 8H, Ar-H), 4.64-4.48 (m, 3.6H, benzyl-H, dioxane-H, pip-H), 4.04 (m, IH, dioxane-H), 3.83 (s, 0.8H5 benzyl-H), 3.75-3.63 (m, 2.4H, dioxane-H, pip-H), 3.56 (s, 1.2H, benzyl-H), 2.92-2.83 (m, 2H, pip-H), 2.44-2.38 (m, 2H, NCH2), 2.09-2.01 (m, 1.2 pip-H), 1.89-1.53 (m, 7H, dioxane-H, pip-H, NCH2CH2), 1.44-1.31 (m, 1.8H, dioxane-H, pip-H), 1.19 (m, 3H, CH3). HPLC tR = 9.0 min.
Example 217 - N-^-Fluorobenzvn^-^-isobutoxyphenvD-N-n-r∑-CΣ.S.S-trimethyl-l.S- dioxan-2-yl)ethyl] piperidin-4-yUacetamide, oxalate π i8AF33-86).
[0697] The desired compound was synthesized from JV-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)-iV-piperidin-4-yl-acetamidel03NLS56 (145 mg, 0.36 mmol) and 2-(2- bromoethyl)-2,5,5-trimethyl-l,3-dioxane (104.5 mg, 0.44 mmol) using the same method as for synthesis of 130AF65-182. Purification of the product by silica gel column chromatography, eluting with 5% methanol in dichloromethane, afforded the title compound (119 mg, 58%).
[0698] R/ = 0.15 (MeOH/CH2Cl2, 5:95). LCMS m/z 555 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.20-6.79 (m, 8H, Ar-H), 4.66-4.56 (m, 0.6H, pip-H), 4.49 & 4.43 (2s, 2H, benzyl-H), 3.76-3.68 (m, 3.2H, pip-H, benzyl-H, CH2OiBu), 3.52-3.47 (m, 4H, dioxane-H), 3.41 (m, 1.2H, benzyl-H), 2.93-2.84 (m, 2H, pip-H), 2.48-2.40 (m, 2H, NCH2), 2.11-2.00 (m, 2.2H, CHOlBu, pip-H),' 1.87-1.80 (m, 2.8H, pip-H, NCH2CH2), 1.72- 1.50 (m, 3.2Η, pip-H), 1.33 (s, 3.8H, CH3, pip-H), 1.02-0.87 (m, 12H, CH3). HPLC JR = 9.8 min.
Example 218 - N-(4-Fluorobenzyiy2-(4-isobutoxyphenyl)-N--fl-f2-f2 -methyl- K3- dioxolan-2-yl)ethyll-Diperidin-4-yl>acetamide, oxalate (118AF35-87).
[0699] The desired compound was synthesized from i\f-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)- iV-piρeridin-4-yl-acetamide 103NLS56 (311 mg, 0.78 mmol) and 2-(2- bromoethyl)-2-methyl-l,3-dioxolane (188 mg, 0.96 mmol) using the same method as for synthesis of 130AF65-182. Purification by silica gel column chromatography, eluting with 5% methanol in dichloromethane, afforded the title compound (61 mg, 15%). [0700] R/ = 0.20 (MeOH/CH2Cl2, 5:95). LCMS m/z 513 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.17-6.76 (m, 8H, Ar-H), 4.64-4.52 (m, 0.6H, ρiρ-H), 4.47 & 4.41 (2s, 2H, benzyl-H), 3.93-3.82 (m, 4H, dioxolane-H), 3.76-3.63 (m, 3.2H, benzyl-H, CH2OiBu, pip-H), 3.47 (s, 1.2H5 benzyl-H), 2.94-2.83 (m, 2H, pip-H), 2.43-2.32 (m, 2H, NCH2), 2.12-1.97 (m, 2.2H, CHOJBU, pip-H), 1.84-1.72 (m, 2.8H, pip-H, NCH2CH2), 1.70- 1.50 (m, 3.2H, ρiρ-H), 1.27 (s, 3.8H, CH3, pip-H), 0.98 (m, 6H, CH3OiBu). HPLC /R= 8.8 min.
Example 219 - N-(4-FluorobenzylV2-(4-isobutoxyphenyl)-N-f l-f3-(1.3-dioxolan-2- yDpropyl~|piperidin-4-vUacetamide, tartrate (118AF79-39").
[0701] The desired compound was synthesized from iV-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)- JV-piperidin-4-yl-acetamide 103NLS56 (156 mg, 0.39 mmol) and 2-(3- chloropropyl)-l,3-dioxolane (62 μL, 0.47 mmol) using the same method as for synthesis of 130AF65-182. Purification by silica gel column chromatography, eluting with a stepwise gradient of 0-4% methanol in dichloromethane, afforded the title compound (49 mg, 25%).
[0702] R/ = 0.45 (MeOH/CH2CI2, 7:93). LCMS m/z 513 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.21-6.79 (m, 8H, Ar-H), 4.84 (t, IH, J = 4.4, dioxolane-H), 4.66-4.56 (m, 0.6H, ρiρ-H), 4.50 & 4.44 (2s, 2H, benzyl-H), 3.95-3.90 (m, 2H, dioxolane- H), 3.84-3.67 (m, 5.2H, benzyl-H, CH2OiBu, pip-H, dioxolane-H), 3.50 (s, 1.2H, benzyl- H), 2.94-2.84 (m, 2H, pip-H), 2.34-2.27 (m, 2H, NCH2), 2.10-1.98 (m, 2.2H, CHOJBU, pip- H), 1.84-1.78 (m, 0.8H, pip-H), 1.71-1.50 (m, 7.2H, pip-H, NCH2CH2), 1.34-1.25 (m, 0.8H, pip-H), 1.01 (m, 6H, CH3OiBu)- HPLC /R= 8.0 min.
Example 220 - N-(4-Fluorobenzyl*)-2-(4-isobutoxyphenylVN-(l-r3-piperidin-l-yl- propyl*)piperidin-4-vU-acetamide, dihydrochloride (9%AF26-4Ti.
[0703] The desired compound was synthesized from iV-(4-fiuorobenzyl)-2-(4- isobutoxyphenyl)- iV-piperidin~4-yl-acetamidelO3NLS56 (189 mg, 0.47 mmol), 1- piperidine (61 μL, 0.61 mmol) and l-chloro-3-iodopropane (61 μL, 0.57 mmol) using the same method as for synthesis of 130AF09-145. Purification of the product by silica gel column chromatography, eluting with 10% methanol in dichloromethane, afforded the title compound (75.6 mg, 31%). [0704] R/ = 0.13 (MeOH/CH2Cl2, 1:4). LCMS m/z 524 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.21-6.81 (m, 8H3 Ar-H), 4.66-4.54 (m, 0.6H, pip-H), 4.51 & 4.45 (2s, 2H5 benzyl-H), 3.78-3.68 (m, 3.2H, benayl-H, CH20JBu, pip-H), 3.52 (s, 1.2H, benzyl-H), 2.93-2.83 (m, 2H, piρ-H), 2.40-2.23 (m, 8H, NCH2), 2.15-1.26 (m, 15H, pip- H3 CH(CH3)2, CH2), 1.02 (m, 6H, CH(CAj)2). HPLC /R= 8.0 min.
Example 221 - N-(4-Fluorobenzvπ-2-('4-isobutoxyphenyl)-N-π-r2-('tetrahvdropyran-2- yloxy)ethyl1-piperidin-4-yl|acetamide. oxalate (98AF41-44).
[0705] The desired compound was synthesized from iV-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)- iV-piperidin-4-yl-acetamide 103NLS56 (185 mg, 0.46 mmol) and 2-(2- chloroethoxy)-tetrahydro-2H-pyran (75 μL, 0.51 mmol) using the same method as for synthesis of 130AF09-145. Purification of the product by silica gel column chromatography, eluting with 4.5% methanol in dichloromethane, afforded the title compound (96 mg, 40%).
[0706] R/ = 0.18 (MeOH/CH2Cl2, 4:96). LCMS m/z 527 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.21-6.78 (m, 8H, Ar-H), 4.67-4.56 (m, 0.6H, pip-H), 4.54 (m, IH, THP), 4.49 & 4.44 (2s, 2H, benzyl-H), 3.86-3.66 (m, 5.2H, benzyl-H, CH2OiBu, pip-H, CHO), 3.58-3.43 (m, 3.2H, benzyl-H, CHO), 3.01-2.89 (m, 2H, pip-H), 2.62 & 2.55 (2t, 2H, J = 6.0, NCiZ2CH2O), 2.26-2.17 (m, 1.2H, pip-H), 2.12-1.96 (m, 1.8H, CHoiβu, pip-H), 1.82-1.44 (m, 9.2H, pip-H, THP)3 1.33-1.26 (m, 0.8H, pip-H), 1.01 (m, 6H, CH(CHi)2). HPLC tR= 7.2 min.
Example 222 - N-C4-FluorobenzylV2-('4-isobutoxyphenylVN-ll-[3-(2-oxo-piperidin-l- yl)propyπpiperidin-4-yl}acetamide C98AF73-64').
[0707] Sodium hydride (60% suspension in oil, 26 mg, 0.65 mmol) was added to a solution of 2-piperidone (54 mg, 0.54 mmol) in dry THF (2 mL) under argon atmosphere. After 15 minutes stirring at rt the reaction mixture was cooled to 00C over 15 minutes. A solution of l-bromo-3-chloropropane (160 μL, 1.62 mmol) was added dropwise to the cold mixture and stirring was continued overnight at rt. The mixture was partitioned between water and ethyl acetate, the organic layer dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue on silica gel column chromatography, eluting with a stepwise gradient of 60-80% ethyl acetate in n-heptane, afforded l-(3-chloropropyl)-piperidin-2-one (33 mg, 35%).
[0708] R/= 0.22 (ethyl acetate/n-heptane 8:2). LCMS m/z 176 [M+H]+. HPLC JR = 1.8 min.
[0709] A solution of l-(3-chloropropyl)-piperidin-2-one (32 mg, 0.18 mmol) in dry DMF (2 mL) was added to a suspension of potassium carbonate (52 mg, 0.38 mmol) and //-(4-fluorobenzyl)-2-(4-isobutoxyphenyl)- iV-piperidin-4-yl-acetamide 103NLS56 (62 mg, 0.15 mmol) in dry DMF (2 mL). After addition of sodium iodide (25 mg, 0.17 mmol) the mixture was stirred overnight at 48°C. Afterwards it was partitioned between water and dichloromethane. The organic layer were dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by reversed phase HPLC (Cis) afforded the desired compound (2.6 mg, 3%).
[0710] R/ = 0.11 (MeOH/CH2Cl2 5:95). LCMS m/z 538 [M+H]+. HPLC tR = 8.2 min.
Example 223 - N-(4-Fluorobenzyl)-2-f4-isobutoxyphenylVN-π-[3-(2-oxo-ρyrrolidin-l- yl)propyl]piperidin-4-yl)acetamide, hydrochloride (98AF76-65).
[0711] The desired compound was synthesized from JV-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)- iV-piperidin-4-yl-acetamide 103NLS56 (107 mg, 0.27 mmol), 2- pyrrolidone and l-bromo-3-chloropropane using the same method as for synthesis of 98AF73-64. Purification of the product by silica gel column chromatography, eluting with a stepwise gradient of 4-8% methanol in dichloromethane, afforded the title compound (15 mg, 11%).
[0712] R/ = 0.39 (MeOH/CH2Cl2 1:9). LCMS m/z 524 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.20-6.80 (m, 8H), 4.65-4.53 (m, 0.6H), 4.50 & 4.44 (2s, 2H), 3.76-3.67 (m, 3.2H), 3.51 (m, 1.2H), 3.34 (t, 2H5 J= 7.2), 3.26 (t, 2H, J= 7.2), 2.95-2.82 (m, 2H), 2.38-2.25 (m, 4H), 2.12-1.96 (m, 4.2H), 1.86-1.56 (m, 6H), 1.29 (m, 0.8H), 1.01 (m. 6H). HPLC tR= 7.6 min.
Example 224 - N-r4-Fluorobenzyl)-2-r4-isobutoxyphenyl)-N-{ l-r3-frRV4-isopropyl-2- oxo-oxazolidin-3-yl)propy_1piperidin-4-yl } acetamide, oxalate (98 AF 100-73).
[0713] Sodium hydride (55% suspension in oil, 144 mg, 3.31 mmol) was added to a solution of (i.)-4-isopropyl-2-oxazolidinone (356 mg, 2.75 mmol) in dry tetrahydrofuran (17 mL) under argon atmosphere. The suspension was stirred for 1 hour at rt, then cooled to 00C and a solution of l-bromo-3-chloropropane in dry tetrahydrofuran (3 mL) was added dropwise. After 48 h stirring at 58°C the mixture was quenched with water. The solvent was removed by evaporation under reduced pressure and the residue partitioned between water and dichloromethane. The organic layer was evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with a mixture of ethyl acetate and n-Heptane (70:30), afforded (4i2)-3-(3-chloropropyl)-4- isopropyloxazolidinon-2-one (401 mg, 71 %).
[0714] A solution of (4i?)-3-(3-chloropropyl)-4-isopropyloxazolidinon-2-one (160 mg, 0.78 mmol) in dry DMF (2 mL) was added to a suspension of potassium carbonate (217 mg, 1.57 mmol) and N-(4-fluorobenzyl)-2-(4-isobutoxyphenyl)- N- piperidin-4-yl-acetamide 103NLS56 (250 mg, 0.63 mmol) in dry DMF (6 mL). After addition of sodium iodide (113 mg, 0.75 mmol) the mixture was stirred overnight at 62°C and partitioned between water and dichloromethane. The organic layer were dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with 5% methanol in dichloromethane, afforded the desired compound (143 mg, 40%).
[0715] R/ = 0.28 (MeOH/CH2Cl2 6:96). LCMS m/z 568 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.20-6.78 (m, 8H, Ar-H), 4.61-4.51 (m, 0.6H, pip-H), 4.48 & 4.42 (2s, 2H, benzyl-H), 4.15 (t, IH, J= 8.8, oxa-H), 4.01 (m, IH, oxa-H), 3.78-3.64 (m, 4.2H, pip-H, benzyl-H, oxa-H, CH2OiBu), 3.48 (m, 2.2H, benzyl-H, CONCHCH2), 2.92- 2.79 (m, 3H, pip-H, CONCHCH2), 2.34-2.22 (m, 2H, NCH2CH2CH2), 2.10-1.96 (m, 3.2H, pip-H, CHiPr, CHoiBu), 1.76-1.50 (m, 6H, pip-H, NCH2CH2), 1.32-1.26 (m, 0.8Η, pip-H), 0.99 (m, 6H, CH3OiBu), 0.81-0.87 (m, 6H, CH31Pr). HPLC fe = 9.1 min.
Example 225 - N-f4-Fluorobenzyl)-2-('4-isobutoxyphenyl*)-N-fl-r3-(2-oxo-oxazolidin-3- yl)proρyl]piperidin-4-yllacetamide. oxalate (98AF94-71").
[0716] The desired compound was synthesized from iV-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)- N-piperidin-4-yl-acetamide 103NLS56 (298 mg, 0.75 mmol), 2- oxazolidoneand l-bromo-3-chloroproρane using the same method as for synthesis of 98AF 100-73. Purification of the product by silica gel column chromatography, eluting with 5% methanol in dichloromethane, afforded the title compound (157 mg, 40%). [0717] R/ = 0.23 (MeOH/CH2Cl2 5:95). LCMS m/z 526 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.20-6.78 (m, 8H5 Ar-H)5 4.61-4.50 (m, 0.6H5 pip-H), 4.48 & 4.42 (2s, 2H, benzyl-H), 4.29-4-24 (m, 2H, oxa-OCH2), 3.78-3.65 (m, 3.2H3 pip-H5 benzyl-H, CH2OiBu)5 3.52-3.48 (m; 3.2H, benzyl-H5 oxa-NCH2), 3.25 (t, 2H, J = 7.2, CONCH2CH2CH2N), 2.89-2.80 (m, 2H, pip-H), 2.33-2.26 (m, 2H, NCH2CH2CH2NCO), 2.09-1.76 (m, 3H, pip-H, CHOIBU), 1.71-1.49 (m, 5.2H, pip-H, NCH2CH2CH2), 1.33-1.27 (m, 0.8H, pip-H), 1.00 (m, 6H, CH3OiBu). HPLC /R = 7.8 min.
Example 226 - (SV4-Methyl-oxazolidin-2-one (118AFl 0-77).
[0718] Triethylamine (0.94 raL, 6.65 mmol) was added dropwise to a solution of L-alaninol (500 mg, 6.65 mmol) and 1,1-carbonyldiimidazole (1.29 g, 7.98 mmol) in dry THF (10 raL) at rt, under argon atmosphere. The reaction mixture was stirred overnight at 600C. The solvent was removed by evaporation under reduced pressure. Purification of the residue by silica gel column chromatography, eluting with 6% methanol in dichloromethane, afforded the desired compound (450 mg, 67%).
[0719] R/ = 0.39 (MeOH/CH2Cl2 6:94). 1H NMR (CDCl3) δ 6.74 (m, IH), 4.45-4.34 (m, IH), 4.98-4.77 (m, 2H), 1.17 (m, 3H).
Example 227 - N-(4-FluorobenzylV2-('4-isobutoxyphenylVN-i'l-r3-('fS')-4-methyl-2-oxo- oxazolidin-3-yDpropylipiperidin-4-yl)acetamide,, tartrate Cl 18AF18-81).
[0720] The desired compound was synthesized from iV-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)- N-piperidin-4-yl-acetamide 103NLS56 (205 mg, 0.52 mmol), (<S>4- methyl-oxazolidin-2-one (118AFl 0-77) and l-bromo-3-chloropropane using the same method as for synthesis of 98AF 100-73. Further purification by acidic ion-exchange SPE cartridge was performed. Yield: 106 mg, 38%.
[0721] Ry = 0.22 (MeOH/CH2Cl2 6:94). LCMS m/z 540 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.20-6.78 (m, 8H, Ar-H), 4.61-4.50 (m, 0.6H, pip-H), 4.48 & 4.42 (2s, 2H5 benzyl-H), 4.34 (m5 IH, oxa-H), 3.84-3.66 (m5 5.2H5 pip-H, benzyl-H, oxa- H, CH2OiBu), 3.49 (s, 1.2H, benzyl-H), 3.42-3.34 (m, IH, CONCH2), 3.09-3.00 (m, IH, CONCH2), 2.92-2.79 (m, 2H, pip-H), 2.33-2.26 (m, 2H, NCH2), 2.10-1.98 (m, 2.2H, pip- H, CHoiBu), 1.86-1.76 (m, 0.8H, pip-H), 1.72-1.48 (m, 5.2H, pip-H, NCH2CH2), 1.29 (m, 0.8H, pip-H), 1.22 (m, 3H5 oxa-GH3), 0.99 (m, 6H, CH3OiBu). HPLC rR= 8.4 min. Example 228 - (SV4-Ethyl-oxazolidin-2-one αi8AF08-76).
[0722] Triethylamine (0.80 mL, 5.74 mmol) was added dropwise to a solution of (»S)-(+)-2-amino-l-butanol (515 mg, 5.77 mmol) and 1,1-carbonyldiimidazole (1.10 g, 6.78 mmol) in dry THF (10 mL) at rt under argon atmosphere. The reaction mixture was stirred overnight at rt. The solvent was removed by evaporation under reduced pressure. Purification of the residue by silica gel column chromatography, eluting with 6% methanol in dichloromethane, afforded the desired compound (485 mg, 73%). R/= 0.42 (MeOH/CH2Cl26:94).
Example 229 - N-f4-Fluorobenzvn-2-r4-isobutoxyτ>henylVN-(l-r3-ffSV4-ethyl-2-oxo- oxazolidin-3-ylVpropyl1piperidin-4-yUacetamide. oxalate Cl 18AF16-80V
[0723] The desired compound was synthesized from iV-(4-fluorobenzyl)-2-(4- isobutoxyphenyl)- N-piperidin-4-yl-acetamide 103NLS56 (202 mg, 0.51 mmol), (S)A- ethyl-oxazolidin-2-one (118AF08-76) and l-bromo-3-chloropropane using the same method as for synthesis of 98AF 100-73. Purification of the product by acidic ion- exchange SPE cartridge afforded the title compound (126 mg, 44%).
[0724] R/ = 0.28 (MeOH/CH2Cl2 6:94). LCMS m/z 554 [M+H]+. 1H NMR (CDCl3, rotamers 0.4:0.6) δ 7.20-6.78 (m, 8H5 Ar-H), 4.61-4.52 (m, 0.6H, pip-H), 4.48 & 4.42 (2s, 2H, benzyl-H), 4.32-4.26 (m, IH, oxa-H), 3.94-3.88 (m, IH, oxa-H). 3.76-3.66 (m, 4.2H, pip-H, benzyl-H, oxa-H, CH2OiBu), 3.49 (s, 1.2H, benzyl-H), 3.46-3.37 (m, IH5 CONCH2), 3.04-2.96 (m, IH, CONCH2), 2.90-2.78 (m, 2H5 pip-H), 2.33-2.24 (m, 2H5 NCH2), 2.11-1.96 (m, 2.2H, pip-H, CHOiBu), 1-82-1.75 (m, 0.8H5 pip-H), 1.74-1.42 (m, 7.2H5 pip-H, NCH2CH2, CH2CH3), 1.29 (m, 0.8H5 pip-H), 1.00 (m5 6H, CH3OiBu), 0.85 (m, 3H, CH2CH5). ΗPLC /R = 8.7 min.
Example 230 - 2-C2-Bromoethyl>)-13-oxothiolane (121JPl I).
[0725] Adapting a procedure by Yamada et. al (Tetrahedron Lett., 1998, 39, 7709-7712), boron trifluoride ether complex (5 mL, 40 mmol) was added dropwise to a mixture of 2-(2-bromoethyl)-l,3-dioxolane (1.45 g, 8.0 mmol) and 2-mercaptoethanol (2.81 mL, 40 mmol) in dichloromethane (15 mL) at rt under Ar atmosphere and stirred at rt overnight. Sat. aq. NaHCO3 (100 mL) was added to the crude mixture, followed by extraction using Et2O (3 x 100 mL), drying (Na24) and evaporation in vacuo. Purification by ^Cugelrohr distillation (90 0C, 1.0 mmHg) afforded 1.08 g of the title compound as a yellow oil. The purity of this material after distillation was 71% (determined by GC analysis) and it was used as such in the alkylation step (121 JP12).
Example 231 - N-f4-Fluorobenzvn-2-f4-isobutoxyphenylVN-(l-r2-fl.3-oxothioIan-2- yl)ethyl]piperidin-4-yl}acetamide, L-tartrate (121 JPl 2).
[0726] The title compound was prepared by the general procedure described above for 103NLS63-F using 103NLS56 (130 mg, 0.33 mmol) and 121JP11 (85 mg, 0.43 mmol) as the alkylating agent. Workup as in 12 IJP 11 followed by vacuum filtration chromatography over silica gel (VFC5 ethyl acetate/n-heptane 0:1-» ethyl acetate/«- heptane l :0-> ethyl acetate/MeOH 4:1) gave 85 mg (51 %) of 121JP12 as colorless thick oil. The Z-tartrate salt was prepared as described above.
[0727] R/ = 0.57 (MeOH/CH2Cl2 1:10). LCMS m/z 515 [M+H]+. 1H-NMR (CDCl3, rotamers 0.5:0.5) δ 7.20-6.76 (m, 8H), 5.10-5.00 (m, IH, oxothiolane-H), 4.66- 4.54 (m, 0.5H5 pip-H)5 4.48 and 4.42 (2s, 2H5 benzyl-H), 4.30-4..22 (m, IH, oxothiolane- H), 3.78-3.64 (m, 4.5H, pip-H, benzyl-H, oxothiolane-H, OCH2OiBu), 3.48 (s5 IH, benzyl- H)5 3.01-2.82 (m5 4H, pip-H, oxothiolane-H), 2.60-2.34 (m, 2H, NCH2), 2.21-1.56 (m, 8H5 pip-H, NCH2CH2, CHoiBu), 1.32-1.22 (m, IH, pip-H), 1.04-0.96 (m, 6H, CH3OiBu- HPLC tR = 10.1 min.
Example 232 - 2-(4-BromophenylVN-π-|"2-(1.3-dioxan-2-vnethvDpiperidin-4-vU-N-(4- fluorobenzylVacetamide, L-tartrate (121JP13).
[0728] The title compound was prepared by the procedure described above for 117NLS87-A using 118AF52-95 (200 mg, 0.62 mmol) and 4-bromophenylacetic acid (500 mg, 2.32 mmol). Sat. aq. NaHCO3 (100 mL) was added to the crude mixture, followed by extraction using CH2CI2 (3 x 100 mL), drying (Na2SO4) and evaporation in vacuo. VFC over silica gel (ethyl acetate/w-heptane 1:1—» ethyl acetate/«-heptane 1 :0-» ethyl acetate/MeOH 2:1) gave 250 mg (78 %) of 121 JP12 as a thick oil. The Z-tartrate salt was prepared as described above.
[0729] R/ = 0.49 (MeOH/CH2Cl2 1:10). LCMS m/z 521 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.50-6.88 (m, 8H), 4.62-4.57 (m, 0.4H, pip-H), 4.50 (t, IH, J = 4.9, dioxane-H) 4.48 and 4.42 (2s, 2H, benzyl-H), 4.06-4.00 (m, 2H, dioxane-H), 3.76 and 3.50 (2s, 2H, benzyl-H), 3.75-3.60 (m, 2.6H5 pip-H, dioxane-H), 3.01 and 2.90 (2d, 2H, J = 10.5, pip-H), 2.52 and 2.41 (2t, 2H, J = 8.0, NCH2), 2.10-1.98 (m, 2.2 H, dioxane- H, pip-H), 1.97-1.58 (m, 6H, pip-H, NCH2CH2), 1.38-1.20 (m, 1.8H, dioxane-H, pip-H). HPLC tR = 8.3 min.
Example 233 - N-U-r2'π.3-Dioxan-2-vnethyl')piperidin-4-vU-N-('4-fluorobenzylV2-('4- isobutylamino-phenvDacetamide, L-tartrate ( 121 JP27").
[0730] Adapting a protocol by Buchwald et al (J. Am. Chem. Soc, 1996, 118, 7215-7216), 12 UPl 3 (100 mg, 192 μmol), isobutylamine (17 mg, 230 μmol), Pd2dba3 (11.6 mg, 19.2 μmol), BINAP (12.0 mg, 38.4 μmol) and NaOtBu (25.8 mg, 269 μmol) were weighed into a flask, toluene (2 mL) was added and the resulting mixture was stirred at 80 0C for 18 h. Workup as in 121 JP 13 followed by preparative reversed-phase (Ci8) HPLC afforded 25.7 mg (27.0 %) of 121JP27 as a thick colorless oil. The Z-tartrate salt was prepared as described above.
[0731] R/ = 0.30 (MeOH/CH2Cl2 1:10). LCMS m/z 512 [M+H]+. 1H-NMR (CDCl3, rotamers 0.5:0.5) δ 7.10-6.81 (m, 6H), 6.59-6.49 (m, 2H), 4.65-4.55 (m, 0.5H, pip-H), 4.55-4.50 (m, IH, dioxane-H), 4.50 and 4.43 (2s, 2H, benzyl-H), 4.10-4.02 (m, 2H, dioxane-H), 3.80-3.67 (m, 3.5H, pip-H, benzyl-H, dioxane-H) 3.45 (s, IH, benzyl-H), 2.95-2.85 (m, 4H, pip-H, NHCH2CH(CH3)2)), 2.45-2.35 (m, 2H, NCH2), 2.09-1.99 (m, 2H, dioxane-H, pip-H), 1.91-1.50 (m, 7H, NCH2CH2, pip-H, NHCH2CH(CH3)2), 1.38- 1.25 (m, 2H, dioxane-H, pip-H), 0.98 (m, 6H, NHCH2CH(CHs)2). HPLC tR = 8.2 min.
Example 234 - N-f l-r2-π3-Dioxan-2-yl')ethvnpiperidin-4-vπ-N-f4-fluorobenzvn-2-(4- propylamino-phenvDacetamide, L-tartrate (121JP28').
■ [0732] Prepared identically as described in the protocol for the synthesis of 121JP27, using propylamine (16 mg, 230 μmol) instead of isobutylamine to afford 24 mg (25 %) of 121JP28 as a thick oil. The Z-tartrate salt was prepared as described above.
[0733] Rf = 0.33 (MeOH/CH2Cl2 1:10). LCMS m/z 498 [M+H]+. 1H-NMR (CDCl3, rotamers 0.5:0.5) δ 7.11-6.82 (m, 6H), 6.53-6.43 (m, 2H), 4.58-4.49 (m, 0.5H, pip-H), 4.48-4.45 (m, IH, dioxane-H), 4.42 and 4.35 (2s, 2H, benzyl-H), 4.05-3.95 (m, 2H, dioxane-H), 3.70-3.60 (m, 3.5H, pip-H, benzyl-H. dioxane-H), 3.40 (s, IH, benzyl- H), 3.05-2.95 (m, 2H, pip-H), 2.85-2.70 (m, 2H, NHCH2CH2CH3), 2.48-2.38 (m, 2H, NCH2), 2.05-1.90 (m, 2H5 dioxane-H, pip-H), 1.92-1.40 (m, 8H5 NCH2CH2, pip-H, NHCH2CH2CH3), 1.40-1.28 (m, 2H5 dioxane-H, pip-H), 0.98 (m, 3H, NHCH2CH2CH3). HPLC IR = 7.3 min.
Example 235 - N-π-r2-π3-Dioxan-2-vnethvnpiperidin-4-vU-N-(4-fluorobenzylV2-('4- (l-nitropropyl)-phenyθacetamide. L-tartrate (121 JP34).
[0734] Adapting a protocol by Vogl & Buchwald (J. Org. Chem., 2002, 67, 106-111), 121JP13 (135 mg, 0.26 mmol), 1-nitroproρane (47 mg, 0.52 mmol), Cs2CO3 (95 mg, 0.29 mmol), 2-di-ter/-butylphosphinobiphenyl (15.5 mg, 52 μmol) and Pd2dba3 (11.9 mg, 13μmol) were weighed into a flask, DME (2 mL) was added and the reaction was stirred at 60 0C for 20 h. Workup as in 12 UP 13 followed by preparative TLC (CH2Cl2/MeOH, 15:1, 10 x eluted) afforded 22 mg (16 %) of 121JP34 as a thick colorless oil. The L-tartrate salt of the title compound was prepared as described above.
[0735] R/ = 0.58 (MeOH/CH2CI2 1 :10). LCMS m/z 528 [M+H]+. HPLC /R =
8.1 min.
Example 236 - N-π-r2-(1.3-Dioxan-2-yl)ethyl)piperidin-4-vU-N-f4-fluoroben2ylV2-r4- (2-oxopyrrolidin-l-y0phenyl)acetamide, L-tartrate ("121JP3 P.
[0736] Adapting a protocol by Yin & Buchwald (J. Am. Chem. Soc, 2002. 124, 6043-6048), 121JP13 (124 mg, 0.24 mmol), pyrrolidone (24.7 mg, 0.29 mmol), Cs2CO3 (111 mg, 0.34 mmol), Xantphos (20.8 mg, 0.036 mmol) and Pd2dba3 (11.0 mg, 0.012 mmol) were weighed into a flask, dioxane (2 mL) was added and the reaction was stirred at 90 0C for 70 h. Workup as in 121 JP13 and purification as in 121JP27 afforded 8 mg (7 %) of 121JP31 as a thick colorless oil. The /--tartrate salt was prepared as described above.
[0737] R/ = 0.31 (MeOH/CH2Cl2 1 :10). LCMS m/z 524 [M+H]+. 1H-NMR (CDCl35 rotamers 0.6:0.4) δ 7.60-6.80 (m, 8H), 4.60-4.50 (m, 0.4H, pip-H), 4.47 (t, IH, J = 5.1, dioxane-H) 4.42 and 4.38 (2s, 2H, benzyl-H), 4.04-3.97 (m, 2H, dioxane-H), 3.82- 3.60 (m, 5.4H, pip-H, dioxane-H, benzyl-H5 pyrrol-H), 3.25 (s, 1.2H, benzyl-H), 2.90- 2.72 (m, 2H5 pip-H), 2.60-2.50 (m, 2H, pyrrol-H), 2.39-2.32 (m, 2H, NCH2) 2.18-1.90 (m,
4.2 H, dioxane-H, pip-H, pyrrol-H), 1.81-1.40 (m, 6H, pip-H, NCH2CH2), 1.32-1.18 (m, 1.8H, dioxane-H, pip-H). HPLC /R = 4.9 min. Example 237 - N-( l-r2-ri.3-Dioxan-2-vnethvnDiperidin-4-vn-N-r4-fluoroben-rylV2-('4- isobutylsulfanyl-phenvOacetamide, L-tartrate (121 JP33\
[0738] Adapting a protocol by Li (J. Org. Chem., 2002, 67, 3643-3650), 121JP13 (120 mg, 0.231 mmol), 2-methyl-l-propanethiol (25 mg, 0.28 mmol), [(/- Bu)2P(OH)]2PdCl2 (11.6 mg, 0.0231 mmol) and NaO/Bu (44 mg, 0.46 mmol) were weighed into a flask, toluene (2 mL) was added and the reaction was stirred at 110 0C for 16 h. Workup as in 12 UP 13 and purification as in 121JP27 afforded 1.7 mg (1.4 %) of 121 JP33 as a thick colorless oil. The Z-tartrate salt of the title compound was prepared as described above.
[0739] R/ = 0.46 (MeOH/CH2Cl2 1:10). LCMS m/z 529 [M-I-H]+. 1H-NMR (CDCl3. rotamers 0.6:0.4) δ 7.24-6.82 (m, 8H), 4.57-4.48 (m, 0.4H, pip-H), 4.47 (t, IH, J = 5.1, dioxane-H), 4.45 and 4.38 (2s, 2H, benzyl-H), 4.05-3.95 (m, 2H, dioxane-H), 3.72 (s, 0.8 H, benzyl-H), 3.70-3.60 (m, 2.6H, piρ-H, dioxane-H) 3.44 (s, 1.2H, benzyl-H), 2.87-2.75 (m, 2H, pip-H), 2.72 (t, 2H, J = 6.5, SCH2CH(CHa)2)), 2.38-2.28 (m, 2H, NCH2), 2.05-1.88 (m,, 2.2H, dioxane-H, pip-H), 1.81-1.48 (m, 7H, NCH2CH2, pip-Η, SCΗ2CH(CΗ3)2), 1.30-1.20 (m, 1.8H, dioxane-H, pip-H), 0.98 (t, 6H, J = 6.7, SCH2CH(CH3)2). HPLC rR = 8.8 min.
Example 238 - N-π-r2-(1.3-Dioxan-2-vnethyl')Diρeridin-4-vU-N-r4-fluorobenzylV2-(4- iodophenvD-acetamide, L-tartrate (121JP40).
[0740] The title compound was prepared by the procedure described above 117NLS87-A using 118AF52-95 (400 mg, 1.24 mmol) and 4-iodophenylacetic acid (1.22 g, 4.64 mmol). Workup as in 121 JPl 3 and purification as in 121JP34 gave 320 mg (46 %) of 121 JP40 as a colorless thick oil. The Z-tartrate salt was prepared as described above.
[0741] Rf = 0.52 (MeOHZCH2Cl2 1:10). LCMS m/z 567 [M-I-H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.65-7.55 (m, 2H), 7.16-6.85 (m, 6H), 4.59-4.50 (m, 0.6H, pip-H), 4.51 (t, IH, J= 5.0, dioxane-H), 4.50 and 4.42 (2s, 2H, benzyl-H), 4.09-4.00 (m, 2H, dioxane-H), 3.75 and 3.4.9 (2s, 2H, benzyl-H), 3.74-3.54 (m, 2.4 H, pip-H, dioxane- H), 2.85 (d, 2H, J= 10.6, pip-H), 2.41-2.35 (m, 2H, NCH2), 2.08-1.95 (m, 2.2H, dioxane- H, pip-H), 1.88-1.50 (m, 6H3 pip-H, NCH2CH2), 1.39-1.27 (m, 1.8H5 dioxane-H, pip-H). HPLC tκ = 8.6 min. Example 239 - 2-(r4-AcetoρhenylVN-(l-f2-π.3-dioxan-2-yl')ethvnpiperidin-4-vU-N-f4- fluorobenzylVacetamide. L-tartrate O21JP44).
[0742] Adapting a protocol by Cacchi et al (Org. Lett, 2003, 5, 289-293), 121JP40 (68 mg, 0.12 mmol), acetic anhydride (61 mg, 0.6 mmol), Pd2dba3 (1.4 mg, 1.5 μmol), lithium chloride (26 mg, 0.6 mmol) and EtNZPr2 (31 mg, 0.24 mmol) were weighed into a flask, DMF (0.9 mL) was added and the resulting mixture was stirred at 100 0C for 18 h. Workup as in 12 UP 13 and purification as in 121JP34 afforded 19 mg (33 %) of 121JP44 as a thick colorless oil. The L-tartrate salt was prepared as described above.
[0743] R/ = 0.50 (MeOH/CH2Cl2 1:10). LCMS m/z 483 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) 7.88-7.78 (m, 2H), 7.36-6.84 (m, 6H), 4.58-4.49 (m. 0.4H, pip- H), 4.48-4.46 (m, IH, dioxane-H), 4.45 and 4.38 (2s, 2H, benzyl-H), 4.05-3.95 (m, 2H, dioxane-H), 3.81 and 3.55 (2s, 2H, benzyl-H), 3.70-3.60 (m, 2.6H, pip-H, dioxane-H) 2.85-2.75 (m, 2H, ρip-H), 2.54 and 2.52 (2s, 3H, CH3), 2.38-2.27 (m, 2H, NCH2), 2.05- 1.92 (m, 2.2H, dioxane-H, pip-H), 1.81-1.45 (m, 6H, NCH2CH2, pip-H), 1.32-1.22 (m, 1.8H, dioxane-H, pip-H). HPLC tR = 5.5 min.
Example 240 - 2-r4-(l-HvdroxyiminoethvDphenyl1-N-π-r2-π3-dioxan-2- y0ethyl)piperidin-4-vU-N-(4-fluorobenzyl)acetamide. L-tartrate (121JP48).
[0744J 121JP44 (14 mg, 29 μmol), pyridine (4.6 mg, 58 μmol) and ethanol (5 mL) were placed in a flask, to which hydroxylamine hydrochloride (4.1 mg, 58 μmol) was added and the resulting mixture was stirred at rt for 5 h. Workup as in 12 UP 13 and purification as in 121JP34 afforded 7 mg (49 %) of 121JP48 as a thick colorless oil. The .L-tartrate salt was prepared as described above.
[0745] R/ = 0.40 (MeOHZCH2Cl2 1:10). LCMS m/z 498 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) 7.63-7.51 (m, 2H), 7.33-6.88 (m, 6H), 4.66-4.58 (m, 0.4H, pip- H), 4.56-4.53 (m, IH5 dioxane-H), 4.51 and 4.40 (2s, 2H, benzyl-H), 4.10-4.04 (m, 2H, dioxane-H), 3.85 and 3.58 (2s, 2H, benzyl-H), 3.78-3.67 (m, 2.6H, pip-H, dioxane-H) 2.97-2.83 (m, 2H, pip-H), 2.47-2.37 (m, 2H, NCH2), 2.26 and 2.24 (2s, 3H, CH3), 2.12- 1.98 (m, 2.2H, dioxane-H, pip-H), 1.88-1.58 (m, 6H, NCH2CH2, pip-H), 1.37-1.29 (m, 1.8H, dioxane-H, pip-H). HPLC /R = 4.0 min. Example 241 - N-π-r2-π.3-Dioxan-2-yl>)ethvnpiperidin-4-yl>-N-(4-fluorobenzylV2-r4- morpholin-4-yl-phenvDacetamide. L-tartrate ("121 JP49*).
[0746] Adapting a protocol by Buchwald et'al (Org. Lett., 2002, 4, 581-584), 121JP40 (50 mg, 88 μmol), morphoiine (9.2 mg, 106 μmol), CuI (1.7 mg, 8.8 μmol) and K3PO4 (37.6 mg, 177 μmol) were weighed into a flask in air atmosphere, ethylene glycol (2 mL) was added and the resulting mixture was stirred at 80 0C for 16 h under air atmosphere. Workup as in 121JP13 and purification as in 121JP34 afforded 4.7 mg (10 %) of 121JP49 as a thick colorless oil. The i-tartrate salt was prepared as described above.
[0747] R/ = 0.33 (MeOH/CH2Cl2 1:10). LCMS m/z 526 [M+H]+.1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.18-6.72 (m, 8H)5 4.62 and 4.37 (2s, 2H, benzyl-H), 4.57- 4.50 (m, 0.4H, pip-H), 4.50-4.42 (m, IH, dioxane-H), 4.05-3.95 (m, 2H, dioxane-H), 3.82-3.75 (m, 4H, morph-H), 3.69 and 3.43 (2s, 2H, benzyl-H), 3.68-3.61 (m, 2.6H, pip- H, dioxane-H), 3.12-3.03 (m, 4H, morph-H), 2.85-2.75 (m, 2H, ρiρ-H), 2.38-2.27 (m, 2H, NCH2), 2.07-1.90 (m, 2.2H, dioxane-H, pip-H), 1.82-1.45 (m, 6H, NCH2CH2, pip-Η), 1.30-1.22 (m, 1.8Η, dioxane-H, pip-H). HPLC tR = 6.2 min.
Example 242 - N-π-f2-ri.3-Dioxan-2-yl)ethyl)piperidin-4-vU-N-('4-fluorobenzylV2-f4- pyrazol-l-yl-phenvDacetamide, L-tartrate (121JP56).
[0748] Adapting a protocol by Buchwald et al (J. Am. Chem. Soc, 2001, 123, 7727-7729), 121JP40 (48 mg, 85 μmol), pyrazole (7 mg, 102 μmol), CuI (0.4 mg, 1.7 μmol), racemic /7vms-l,2-cyclohexanediamine (1.0 mg, 8.5 μmol), and K.2CO3 (25 mg, 181 μmol) were weighed into a flask, dioxane (1.5 mL) was added and the resulting mixture was stirred at 110 0C for 60 h. Workup as in 12 UPl 3 and purification as in 121 JP34 afforded 3.9 mg (9 %) of 121 JP56 as a thick colorless oil. The /.-tartrate salt was prepared as described above.
[0749] R/ = 0.27 (MeOH/CH2Cl2 1 :10). LCMS m/z 507 [M+H]+. 1H-NMR (CDCl3, rotamers 0.6:0.4) δ 7.86 and 7.82 (2d, IH, J= 2.2, pyraz-H), 7.64 (d, IH, J= 4.4, pyraz-H), 7.62-6.83 (m, 8H), 6.42-6.36 (m, IH, pyraz-H), 4.60-4.49 (m, 0.6H, ρiρ-H), 4.48 (t, IH, J = 5.1, dioxane-H), 4.45 and 4.38 (2s, 2H, benzyl-H), 4.05-3.95 (m, 2H, dioxane-H), 3.80 and 3.54 (2s, 2H, benzyl-H), 3.70-3.61 (m, '2.4H, pip-H, dioxane-H) 2.85-2.75 (m, 2H5 pip-H), 2.38-2.28 (m, 2H, NCH2), 2.07-1.90 (m, 2.2H5 dioxane-H5 pip- H)5 1.82-1.45 (m, 6H5 NCH2CH2, pip-H)5 1.35-1.22 (m5 1.8H5 dioxane-H, pip-H). HPLC ^R = 6.4 min.
Example 243 - 2-f2-BromopropyD-1.3-dioxane f!21 JP80X
[0750] Adapting a procedure of Buchi and Wuest (J; Org. Chem, 1969, 34, 1122-1123), crotonaldehyde (3 g, 43 mmol) was added dropwise to cone. aq. HBr (5.2 g, 64 mmol) over 5 min at 5 0C under air atmosphere. After 15 min of stirring at 5 0C5 during which time the mixture changed from colorless to brownish, 1,3 -propanediol (8.1 g, 107 mmol) was added and the reaction was stirred at 5 0C for further 0.5 h before allowing it warm to rt5 and finally stirring it at rt for 2 h. The crude reaction mixture was then «-heptane extracted (2 x 200 mL), the combined «-heptane extracts were Na2SO4 dried, evaporated in vacuo and 121JP80 was isolated by Kugelrohr distillation (75 0C5 0.18 mmHg) to obtain 124 mg (1.4 %) of the title compound as a colorless liquid.
[0751J Characterization Data: 1H-NMR (CDCl3) δ 4.73 (dd, IH, J= 7.2, 3.4), 4.26-4.18 (m, IH), 4.15-4.06 (m, 2H)5 3.83-3.75 (m, 2H), 2.15-1.97 (m, 3H), 1.71 (d, 3H, J = 6.6), 1.38-1.32 (m, IH).
Example 244 - N-(l-r2-π3-Dioxan-2-ylVl-methylethvnpiperidin-4-vU-N-f4- fluorobenzyl)-2-(4-iso-butoxyphenyl)-acetamide. L-tartrate (121 JP84).
[0752] The title compound was prepared by the procedure described above 103NLS63-F using 103NLS56 (202 mg, 0.51 mmol) and 121JP80 (124 mg, 0.59 mmol) as the alkylating agent. Workup as in 121 JPl 3 and purification as in 121JP34 gave 1.9 mg (0.7 %) of 121 JP84 as a thick oil. The Z-tartrate salt was prepared as described above.
[0753] R/ = 0.43 (MeOH/CH2Cl2 1:10). LCMS m/z 527 [M+H]+. 1H-NMR (CDCl3, rotamers 0.5:0.5) δ 7.19-6.80 (m, 8H)5 4.50-4.33 (m, 3.5H5 dioxane-H, benzyl-H, pip-H), 4.04-3.93 (m, 2H5 dioxane-H), 3.72-3.55 (m, 5.5H5 dioxane-H, benzyl-H, pip-H, OCH2OiBu), 3.42 (s, IH, benzyl-H), 2.78-2.59 (m, 2H, piρ-H), 2.34-2.16 (m, IH, NCH)5 2.08-1.89 (m, 2H, pip-H, CHOJBU), 1-79-0.77 (m, 17H, CH3OiBu, NCHCH3, NCHCH2, pip- Η, dioxane-Η). ΗPLC tR = 8.5 min.
Example 245 - 4-Iodophenylacetic acid ethyl ester Cl 21 JP58'). [0754] 4-Iodophenylacetic acid (3 g), ethanol (20 mL) and cone. H2SO4 (5 mL) were refluxed overnight. Ca. 15 mL ethanol was then evaporated, the residue was extracted with dichloromethane (3 x 100 mL), the combined organic extracts were washed with sat. aq. NaHCO3, dried over Na2SO4 and evaporated in vacuo to afford 2.97 g (90 %) of 121 JP58 as a yellow oil.
[0755] Characterization Data: 1H-NMR (CDCl3) δ 7.62 (d, 2H, J = 8.4), 7.02 (d, 2H, J= 8.4), 4.07 (q, 2 H, J= 7.0), 3.59 (s, 2H), 1.12 (t, 3H, J= 7.0).
Example 246 - Pyrazol-1-ylphenylacetic acid ethyl ester (121JP64).
[0756] 121 JP58 (290 mg, 1.0 mmol) was treated identically as 121 JP40 for the synthesis of 121JP56. After heating the reaction to 110 0C for 72 h and workup as in 12 UP 13, the crude mixture was purified by VFC (CH2Cl2/MeOH 1 :0 → 20:1) to furnish 180 mg (78 %) of 121 JP64 as a yellow oil.
[0757] Characterization Data: 1H-NMR (CDCl3) δ 7.92 (dd, IH, J= 2.3, 1.0), 7.72 (d, IH, J = 1.3), 7.62 (d, 2H, J = 8.7), 7.39 (d, 2H, J = 8.7), 6.42 (dd, IH, J= 2.5, 1.9), 4.18 (q, 2 H, J= 7.0), 3.61 (s, 2H), 1.22 (t, 3H, J= 7.1).
Example 247 - 4-Pyrazol-l-vIphenylacetic acid (12UP68. 87).
[0758] 12UP64 (180 mg, 0.78 mmol), lithium hydroxide monohydrate (164 mg, 3.9 mmol), H2O (10 mL) and THF (10 mL) were stirred overnight at rt. The crude mixture was then extracted with dichloromethane (3 x 150 mL), the pH of the aqueous phase was adjusted to ca. pH 3 using 4M HCl and extracted with dichloromethane (3 x 150 mL). The combined organic layers were dried over Na2SO4, filtered and evaporated in vacuo to provide 128 mg (81 %) of 121 JP68 as a yellow solid.
[0759] Characterization Data: 1H-NMR (CDCl3) δ 7.90 (m, IH), 7.75 (m, IH), 7.63 (d, 2H, J= 8.6), 7.38 (d, 2H, J= 8.6), 6.45 (m, IH), 3.68 (s, 2H).
Example 248 - N-ll-r2-π.3-Dioxan-4-vnethyl)piperidin-4-yl>-N-r4-fluorobenzvn-2-(4- pyrazol-l-yl-phenvDacetamide, L-tartrate (12UP91).
[0760] The title compound was prepared by the general procedure described above 117NLS87-A using 128NLS52 (87 mg, 0.27 mmol) and 12UP87 (60 mg, 0.27 mmol). Workup as in 12 UP 13 and purification as in 121JP34 gave 25 mg (18 %) of 121 JP91 as a colorless oil. The Z-tartrate salt was prepared as described above.
[0761] R/ = 0.34 (MeOH/CH2Cl2 1 :10). LCMS m/z 507 [M+H]+. 1H-NMR (CDCl3, rotamers 0.5:0.5) δ 7.92 and 7.88 (2d, IH5 J= 2.2, pyraz-H), 7.71 (d, IH, J= 4.7, pyraz-H), 7.69-6.90 (m, 8H), 6.48-6.42 (m, IH, pyraz-H), 5.00 (d, IH, J = 6.3, dioxane- H), 4.65 (d, IH, J = 6.4, dioxane-H), 4.63-4.55 (m5 0.5H, pip-H), 4.52 and 4.46 (2s, 2H, benzyl-H), 4.10-4.02 (m, IH5 dioxane-H), 3.86 and 3.57 (2s, 2H, benzyl-H), 3.78-3.55 (m, 2.5H, pip-H, dioxane-H) 2.93-2.82 (m, 2H, pip-H), 2.49-2.30 (m, 2H, NCH2), 2.10-1.98 (m, IH, pip-H), 1.90-1.33 (m, 9H, NCH2CH2, pip-H, dioxane-H). HPLC /R = 5.2 min.
Example 249 - N-ri-((RV3.5-Dihvdroxypentvnpiperidine-4-vn-N-r4-fluorobenzylV2-(4- isobutoxyphenyPacetamide. tartrate (130AF93-189).
[0762] The desired compound was synthesized from (>S)-5-[(4- methylbenzenesulfonyl)oxy] pentane-l,3-diol (Moune et al, J.Org.Chem., 1997, 62, 3332- 3339) and 103NLS56 using the same method as described for the preparation of 130AF65-182.
[0763] R/= 0.48 (MeOHZCH2Cl2, 10:90). LCMS m/z 501 [M+H]+. HPLC /R = 7.4 min.
Example 250 - N-π-r2-(rf4RV1.3-Dioxane-4-yl')e1hyl1piperidine-4-yl>-N-f4- fluorobenzylV2-('4-isobutoxyphenyl>acetamide. tartrate (130AFgS-IgQ).
[0764] The desired compound (7.9 mg, 55%) was synthesized from 130AF93- 189 (18.6 mg, 0.028 mmol) using the same method as for the synthesis of 130AF67-183. The enantiomeric excess was determined to be 99% using chiral HPLC analysis (Chiralpak AD column, 4.6 x 250 mm; heptane/I-PrOH 50:50, 0.3% DEA; 0.5 mL/min; tR 22.7 min). The 1H NMR and LCMS data were identical with 130AF67-183.
Example 251 - 4-(l,2,4-Triazol-4-yl)phenylacetic acid (141JPOlV
[0765] Adapting a protocol by Catarzi et al (J. Med Chem., 2001, 44, 3157- 3165), diformylhydrazine (352 mg, 4.0 mmol) and then dropwise trimethylsilyl chloride (2.53 mL, 20 mmol) and Et3N (1.30 mL, 9.3 mmol) were added to a suspension of 4- aminophenylacetic acid (201 mg, 1.33 mmol) in anhydrous pyridine. The mixture was heated at 100 0C overnight, volatiles were removed at reduced pressure and the resulting solid was treated with water (6 mL), collected, washed with H2O and dried in vacuo to provide 251 mg (93%) of 141JP01 as a light brown solid. LCMS m/z 204[M+H]+. 1H- NMR (DMSO-rfe) δ 9.05 (s, 2H), 7.62 (d, IH5 J= 8.6), 7.40 (d, IH, J= 8.2), 3.61 (s, 2H).
Example 252 - N-f l-r2-π.3-Dioxan-2-vnethyl1piperidin-4-yl>-N-r4-fluorobenzvn-2- r4π.2.4-triazol-4-vnphenvnacetamide. L-tartrate Cl 4 UPl 3\
[0766] The acid 141 JPOl (35 mg, 0.17 mmol), N-{l-[2-(l,3-dioxan-2- yl)ethyl]piperidin-4-yl}-iV:-(4-fluoroben2yl)amine (118AF52-95, 55 mg, 0.17 mmol) and diisopropylethylamine (52 mg, 0.51 mmol) were dissolved in DMF (5 mL). Bromo-tris- pyrrolidino-phosphonium hexafluorophosphate (PyBroP, 119 mg, 0.25 mmol) was added, and the mixture was stirred at rt for 2 h. The mixture was concentrated and passed onto an • acidic ion exchange SPE cartridge. The cartridge was washed with methanol (8x4 mL) and the remaining product was eluted off the column with 10% NH4OH in methanol (2x4 mL) and evaporated. The resulting oil was purified as in 121JP34 to give 47 mg (54 %) of 141 JPl 3 as a colorless oil. The /.-tartrate salt was prepared as described above.
[0767] R/ = 0.26 (MeOH/CH2Cl2 1 :10). LCMS m/z 508[M+H]+. 1H-NMR' (CDCl3, rotamers 0.5:0.5) δ 8.47 and 8.41 (2s, IH, -H), 7.48-6.89 (m, 8H3 Ar-H), 4.62- 4.56 (m, 0.6H, pip-H), 4.56-4.49 (m, 3H, dioxane-H, benzyl-H), 4.10-4.01 (m, 2H, dioxane-H), 3.79 and 3.61 (2s, 2H, benzyϊ-H), 3.77-3.67 (m, 2.4H, pip-H, dioxane-H), 2.94-2.84 (m, 2H, pip-H), 2.45-2.35 (m, 2H, NCH2), 2.10-1.43 (m, 9H, dioxane-H, NCH2CH2, pip-Η), 1.37-1.27 (m, 1 Η, dioxane-Η).
Pharmalogical Data
Example 253 - Receptor Selection and Amplification (R-SAT) Assays.
[0768] The functional receptor assay, Receptor Selection and Amplification
Technology (R-SAT™), was used (with minor modifications from the procedure described previously (Brann, M. R. US Patent 5,707,798, 1998; Chem. Abstr. 1998, 128,
111548) to screen compounds for efficacy at the 5-ΗT2A receptor. The RSAT assay was conducated as described herein. The results are summarized below in Table 5. Table 5. Efficiency and pIC50 of Compounds at the 5-HT2A Receptor Compared to Ritanserin.
Figure imgf000212_0001
Example 254 - In vivo determination of behavioral effects Animals and Apparatus.
[0769] Male NSA mice (Harlan; San Deigo, CA) were used as subjects. Mice weighed 20 — 30 g. Animals were housed 8/cage in the One Cage system (One Cage; Lab Products, Inc., Seaford, DE) with bedding (1/8 inch Bed "O" Cob; Harlan Teklad, Madison, WI) in a room with controlled temperature 22 ± 30C and a 12 hour lightrdark cycle (lights on 6 am). Water and standard rodent chow (Harlan Teklad) were continuously available in the home cage. For testing, plastic locomotor activity cages (20 x 20 x 30 cm; AccuScan Instruments, Columbus, OH) were equipped with photocell beams for monitoring horizontal activity. Data were collected using Versamax computer software (AccuScan Instruments).
Procedure.
[0770] For determination of spontaneous activity, test compounds were administered alone (s.c. 10 min or p.o. 30 min before the session). For hyperactivity experiments, mice were injected with 0.3 mg/kg MK-801 i.p. 15 min presession (the peak dose for producing hyperactivity in an inverted-U dose-effect curve as determined in pilot experiments) in combination with vehicle or test compound. Motor activity data were collected during a 15 min session in a lit room. Mice had no prior exposure to the motor cages. Each dose or dose combination was tested in a separate group of mice (n=8).
Data Analysis.
[0771] Distance traveled (cm) was calculated and averaged across animals in a group. An analysis of variance (ANOVA) and post-hoc Dunnett's t-test comparions to vehicle control were conducted for each dose-response function. The lowest dose found to be significantly different from vehicle control was defined as the minimum effective dose (MED).
General LC-MS procedure for Example 255 to 287.
[0772] HPLC/MS analyses were performed using either of two general methods (Method A or Method B). Method A: Agilent HPI 100 HPLC/MSD. [0773] G1312A binary pump, G1313A autosampler, G1316A column compartment, G1315A diode array detector (190-450 urn), 1946A MSD, electrospray ionization.
[0774] Chromatography: mobile phase: 8 mM ammoniumacetate in water/acetonitrile. Gradient start at 70% org. up to 100 % org. over 12 min, down to 70 % org. over 0.5 min, held for 3.5 min. Total runtime 16 min. Flowrate 1 mL/min
[0775] Column: Phenomenex Luna Cig(2) 3 μm, 75 x 4.6mm.
[0776] MS parameters: Drying gas, 10 L/min. Nebulizer pressure, 40 psig. Gas temp, 3500C. VCap, 4000. Method B: Waters/Micromass HPLC/MS.
[0777] 600 LC-pump, 2700 sample manager, 996 diode array detector (190- 450 nm), Micromass ZMD-mass-spectrometer, electrospray ionization.
[0778] Chromatography: mobile phase: 10 mM ammoniumacetate in water/acetonitrile. Gradient start at 30% org. for 0.5 min, up to 100 % org. over 9.5 min, held for 2 min, down to 30 % org. over 0.5 min, held for 5.5 min. Total run time 18 min. Flowrate, 1 mL/min.
[0779] Column: Phenomenex Luna Ci8(2) 3 μm, 75 x 4.6mm.
[0780] MS parameters: Desolvation gas, 404 L/h. Capillary, 5.3 kV. Cone, 36 V. Extractor, 3 V. Source block temp, 13O0C. Desolvation temp, 25O0C.
Example 255 - 4-(4-Fluorobenzviy3-(4-isobutoxybenzv0-8-methyl-l-oxa-3,8-diaza- spiror4.5]decan-2-one, hydrochloride T69NLS75*)
[0781] Preparation of 4-[l-Carboxy-2-(4-fluorophenyl)-ethyl]-4-hydroxy-l- methyl-piperidine- (69NLS42).
[0782] n-Butyllithium (1.6 M in hexane, 18.8 mL, 30 mmol) was added dropwise under stirring over 5 min to a cooled solution of N,iV-diisopropylamine (4.2 mL, 30 mmol) in THF (25 mL) at -4O0C. The solution was warmed to O0C and a solution of 3- (4-fluorophenyl)propionic acid (2.53 g, 15 mmol) in THF (20 mL) was added. The mixture was stirred for 30 min at rt, cooled to -78°C and JV-methylpiperidone (2.2 mL, 18 mmol) added dropwise over 15 min. The solution was allowed to warm to rt and poured into a stirred mixture of diethylether (100 mL) and water (60 mL). The organic layer was discarded, the aqueous layer extracted with another portion of diethylether. The aqueous layer was acidified with 4 M aq. HCl and extracted once with dichloromethane and with H-butanol (3x100 mL). The combined n-butanol-extracts were concentrated in vacuo, giving 69NLS42 as a light-yellow oil, which is used without further purification.
[0783] R/- 0.13 (MeOH/CH2Cl2 1 :9).
[0784] Preparation of 4-(4-Fluorobenzyl)-8-methyl-l-oxa-3,8-diaza- spiro[4.5]decan-2-one (69NLS44).
[0785] The crude piperidine derivative 69NLS42 (ca. 15 mmol) was dissolved in toluene (200 mL), triethylamine (4.18 mL, 30 mmol) and diphenylphosphoryl azide (3.88 mL, 18 mmol) were added, and the mixture refluxed overnight. The solvent was removed in vacuo, the residue dissolved in dichloromethane (200 mL) and extracted with IM HCl (3x100 mL). The combined aqueous extracts were basified with 20% aq. KOH solution and extracted with dichloromethane (3x150 mL). The combined organic layers were dried over Na2SO-I, filtered and evaporated to give 69NLS44 (2.06 g, 49% over two steps) as a yellow solid, which was used without further purification.
[0786] R/= 0.25 (MeOH/CH2Cl2 1 :9). LCMS m/z 278 [M+H]+. HPLC tκ = \.9 min (method B).
[0787] Preparation of 4-isobutoxy benzyl bromide (69NLS69):
[0788] The alkylating agent was obtained according to literature procedures from p-cresol by Williamson ether synthesis, followed by a Wohl-Ziegler bromination.
[0789] 4-(4-Fluorobenzyl)-3-(4-isobutoxybenzyl)-8-methyl- 1 -oxa-3,8-diaza- spiro[4.5]decan-2-one, hydrochloride (69NLS75)
[0790] The oxazolidinone 69NLS44 (crude, ca. 7 mmol) was dissolved in DMF/THF (1:9, 50 mL), sodium hydride (50% in oil, 0.67 g, 14 mmol) added and the suspension stirred at rt for 30 min, before the dropwise addition of the bromide 69NLS69 (1.5 g, 7 mmol). The mixture was stirred at rt for 4h and then partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer extracted twice with ethyl acetate. The combined organic extracts were dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography on silica gel, eluting with a stepwise gradient of 0-6% methanol in dichloromethane. Repurification of the pooled fractions on an acidic ion-exchange SPE cartridge afforded the free amine of the title compound 69NLS75 (1.10 g, 34%) as a colorless oil. The compound was dissolved in dichloromethane, treated with an excess of 2 M HCl in diethylether, precipitated from H-heptane, to afford the hydrochloride salt as a colorless solid in quantitative yield.
[0791] R/ = 0.39 (MeOH/CH2Cl2 1:9). LCMS m/z 441 [M+H]+. 1H NMR (CDCl3, free amine) δ 0.96 (d, 6H, J= 6.8, CH3), 1.24-1.32 (m, IH, pip-H), 1.50 (m, IH, pip-H), 1.62-1.70 (m, IH, pip-H), i.83 (m, IH, pip-H), \ .96-2.06 (m, IH, GH(CH3);.), 2.17 (s, 3H5 NCH3), 2.26-2.34 (m5 2H5 pip-H), 2.46 (m, IH, pip-H), 2.60 (m, IH, pip-H), 2.70 (dd, IH, J= 7.3, 14.2, CH2ArF), 2.85 (dd, IH5 J= 6.6, 14.2, CH2ArF), 3.34 (t, IH, J = 7.0, H-4), 3.57 (d, IH5 J= 15.2, CH2ArOiBu), 3.63 (d5 2H5 J= 6.6, OCH2CH), 4.69 (d, IH5 J = 15.2, CH2Ar0JBu), 6.73-6.97 (m, 8H5 Ar-H). 13C NMR (CDCl3) δ 19.5, 28.5, 31.3, 34.2, 36.8, 46.1, 46.3, 51.2, 51.3, 63.5, 74.7, 79.6, 114.9, 116.0 (d, JC-F = 21.0), 127.7, 129.6, 130.6 (d, JC-F = 7.7), 132.9 (d, Jc-F = 3.4), 157.3, 159.2, 162.0 (d, Jc-F = 244.0). HPLC fR = 10.4 min (method B).
Example 256 - 3-(4-Cyclopropylmethoxybenzyl)-4-('4-fluoroben2yl)-8-methyl-l-oxa-3,8- diaza-spiro|"4.51decan-2-one. hydrochloride (69NLS52)
[0792] The title compound was obtained by a procedure similar to the one described for 69NLS75, from 69NLS44 and the appropriate benzylbromide derivative.
[0793] Alternatively, 69NLS44 can be first alkylated with 4-acetoxy benzylbromide, followed by basic treatment and alkylation of the resulting free hydroxyl function with cyclopropylmethyl bromide.
[0794] R/ = 0.45 (MeOH/CH2Cl2 1:9). LCMS m/z 439 [M+H]+. 1H NMR (CD3OD5 free amine) δ 0.33 and 0.60 (2m, 4H5 CH(CH2)2), 1-20-1.26 (m, IH3 CH(CH2)2), 1.37-1.45 (m, IH5 pip-H), 1.58 (m, IH, pip-H), 1.79-1.86 (m, IH5 pip-H), 1.98 (m, IH, pip-H), 2.26 (s, 3H, NCH3), 2.34-2.42 (m, 2H, pip-H), 2.63 (m, IH, pip-H), 2.77 (m, IH5 pip-H), 2.87 (dd, IH5 J= 7.0, 14.2, CH2ArF), 2.98 (dd, IH5 J= 7.0, 14.2, CH2ArF), 3.58 (t, IH, J= 7.0, H-4), 3.72 (d, IH, J = 15.2, CH2ArOMeCpr), 3.80 (d, 2H, J = 6.8, OCH2), 4.61 (d, 1Η, J= 15.2, CΗ2ArOMecPr), 6.84-7.21 (m5 8H, Ar-H). HPLC /R = 6.5 min (method A).
Example 257 - 4-r4-Fluorobenzyl)-3-('4-isobutoxyben2ylV2-oxo-l-oxa-3.8-diaza- spirof4.5]decane-8-carboxylic acid tert-butyl ester (69NLS77)
[0795] Preparation of 4-[l-Carboxy-2-(4-fluoro-phenyl)-ethyl]-4-hydroxy- piperidine-1-carboxylic acid tert-butyl ester (69NLS56). [0796] n-Butyllithium (1.6 M in hexane, 8.8 mL, 14.0 mmol) was added dropwise under stirring over 5 min to a cooled solution of N.N-diisopropylamine (2.0 mL, 14.0 mmol) in THF (10 mL) at -4O0C. The solution was warmed to O0C and a solution of 3-(4-fluorophenyl)propionic acid (1.18 g, 7.0 mmol) in THF (8 mL) was added. The mixture was stirred for 30 min at rt, cooled to -78°C and N-BOC-4-piperidone (1.68 g, 8.4 mmol) in THF (7 mL) added dropwise over 15 min. The solution was allowed to warm to rt and poured into a stirred mixture of diethylether (100 mL) and water (50 mL). The organic layer was discarded, the aqueous layer extracted with another portion of diethylether. The aqueous layer was acidified with 2 M aq. HCl to pH 3.5 and extracted with dichloromethane (3x100 mL). The combined extracts were dried over Na2SC^, filtered and concentrated in vacuo, giving 69NLS56 as a yellow solid, which is used without further purification.
[0797] Rf = 0.42 (MeOH/CH2Cl2 1 :19). LCMS m/z 268 [M-BOC+2H]+. HPLC tR = 3.7 min (method B).
[0798] Preparation of 4-(4-fiuorobenzyl)-2-oxo-l-oxa-3,8-diaza- spiro[4.5]decane-8-carboxylic acid tert-butyl ester (69NLS58).
[0799] The crude piperidine derivative 69NLS56 (ca. 7 mmol) was dissolved in toluene (100 mL), triethylamine (2.0 mL, 14.0 mmol) and diphenylphosphoryl azide (1.8 mL, 14.0 mmol) were added, and the mixture refluxed overnight. The solvent was removed in vacuo, the residue dissolved in ethyl acetate (200 mL) and washed with water and brine. The organic layer was dried over Na2SO4, filtered and evaporated to give 69NLS58. Purification of the residue on silica gel column chromatography, eluting with a stepwise gradient of 0-2% methanol in dichloromethane, afforded 69NLS58 as a yellow solid (1.47 g, 58% overall yield).
[0800] Rf = 0.63 (MeOH/CH2Cl2 1: 19). Rf = 0.33 (ethyl acetate/n-heptane 1:1). LCMS m/z 265 [M-BOC+2H]+. HPLC tR = 10.3 min (method B).
[0801] Preparation of 4-(4-Fluorobenzyl)-3-(4-isobutoxybenzyl)-2-oxo-l-oxa- 3,8-diaza-spiro[4.5]decane-8-carboxylic acid tert-butyl ester (69NLS77).
[0802] The title compound was obtained by alkylation of 4-(4-fluorobenzyl)- 2-oxo-l-oxa-3,8-diaza-spiro[4.5]decane-8-carboxylic acid tert-butyl ester 69NLS58(7.40 g, 20.3 mmol) with 4-isobutoxybenzylbromide (4.88 g, 20.3 mmol) following the same procedure as described for 69NLS75. After purification of the crude product on silica gel column chromatography, using a stepwise gradient of 0-60% ethyl acetate in n-heptane, the collected compound was dissolved in a minimum amount of ethyl acetate and precipitated from n-heptane, giving after filtration 69NLS77 (5.86 g, 55%) as a colorless solid.
[0803J Rf = 0.64 (ethyl acetate/n-heptane 1 :1). LCMS m/z 544 [M+NH4]+. 1H NMR (CDCl3) δ 1.02 (d, 6H, J = 6.6, CH3), 1.36-1.60 (m, 12H, C(CH3)3, ρip-H), 1.88 (m, IH3 pip-H), 2.05-2.11 (m, IH, CH(CH3)2), 2.74 (dd, IH, J = 7.6, 14.2, CH2ArF), 2.95 (dd, IH, J = 6.6, 14.2, CH2ArF), 3.08 (m, 2H, pip-H), 3.39 (t, IH, J = 7.2, H-4), 3.66 (d, IH, J = 15.0, CH2ArOiBu), 3.71 (d, 2H, J = 6.6, OCH2CH), 3.82 (m, IH, pip-H), 3.92 (m, IH, pip-H), 4.77 (d, IH, J = 15.0, CH2ArOiBu), 6.82-7.03 (m, 8H, Ar-H). HPLC tR = 14.8 min (method B).
Example 258 - 8-f2-ri31Dioxolan-2-yl-emyiy4-(4-fluorobenzylV3-f4-isobutoxybenzylV l-oxa-3.8-diaza-spiror4.51decan-2-one, hydrochloride (69NLS79-II)
[0804J 69NLS77 (300 mg, 0.57 mmol) was N-BOC deprotected by treatment with a solution of TFA (2 πiL) in dichlormethane (2 mL) at rt for 1.5 h. The solvent was removed in vacuo, the residue coevaporated twice with.acetonitrile and redissolved in the same solvent (10 mL). Potassium carbonate (110 mg, 0.80 mmol) and 2-(2-bromoethyl)- 1,3-dioxolane (80 μL, 0.68 mmol) were added, followed by sodium iodide (102 mg, 0.68 mmol) and the mixture stirred for 3 days at 5O0C. Partitioning of the mixture between water and dichloromethane, extraction of the aqueous layer twice with dichloromethane, drying of the combined organic layers over Na2SO4, filtering and evaporation of the solvent gave crude 69NLS79-II. The residue was purified by silica gel column chromatography, eluting with a stepwise gradient of 0-4% methanol in dichloromethane, affording 69NLS79-II (127 mg, 0.24 mmol) as a colorless oil. The compound was converted to its HCl form by treatment with 2M HCl in diethylether as described above for 69NLS75, giving the salt as a colorless powder.
[0805] R/ = 0.61 (MeOH/CH2Cl2 1:9). LCMS m/z 527 [M+H]+. 1H NMR (CD3OD, free amine) δ 1.00 (d, 6H, J= 6.6, CH3),.1.34-1.42 (m, IH, pip-H), 1.56 (m, IH, pip-H), 1.72-1.81 (m, 3H, pip-H, O2CHCH2), 1.92 (m, IH, pip-H), 1.97-2.06 (m, IH, CH(CH3)2), 2.27-2.35 (m, IH, pip-H), 2.44 (m, 2H, NCH2), 2.63 (m, IH, pip-H), 2.76 (m, IH, pip-H), 2.85 (dd, IH, J = 7.0, 14.2, CH2ArF), 2.95 (dd, IH, J = 7.0, 14.2, CH2ArF), 3.60 (m, IH, H-4), 3.69 (d, 2H3 J = 6.5, OCH2CH), 3.70 (d, IH, J = 15.2, CH2Ar0JBu), 3.76-3.82 and 3.85-3.91 (2m, 4H, OCH2), 4.60 (d, IH3 J= 15.2, CH2ArOiBu), 4.82 (t, IH, J = 4.7, O2CH), 6.83-7.19 (m, 8H5 Ar-H). HPLC ZR = 11.3 min (method B).
Example 259 - 4-(4-Fluorobenzyl)-3-('4-isobutoxybenzvπ-8-(3-morpholin-4-yl-propyl)-l- oxa-3,8-diaza-spiror4.51decan-2-one, dihydrochloride (69NLS83)
[0806] 69NLS77 (300 mg, 0.57 mmol) was N-BOC deprotected as described in the preparation of 69NLS79-II and dissolved in acetonitrile (3 mL) and DMF (1 mL). To a solution of morpholine (65 μL, 0.74 mmol) in acetonitrile (3 mL) and DMF (1 mL) was added dropwise l-chloro-3-iodopropane (73 μL, 0.68 mmol) and potassium carbonate (300 mg, 2.17 mmol). The mixture was heated at 5O0C for 3 h before the addition of the solution containing the deprotected spiropiperidine followed by sodium iodide (102 mg, 0.68 mmol). The mixture was stirred overnight at 5O0C and worked up as described for 69NLS79-II. Purification of the residue by silica gel column chromatography, eluting with a stepwise gradient of 0-6% methanol in dichloromethane, afforded the desired compound (127 mg, 40%) as a colorless oil. Treatment of the product in dichloromethane with 2 M HCl in diethylether as described for 69NLS75 gave the corresponding dihydrochloride salt as a colorless solid.
[0807] Rr = 0.48 (MeOH/CH2Cl2 1:9). LCMS m/z 554 [M+H]+. 1H NMR (CD3OD5 free amine) δ 1.01 (d, 6H, J = 6.9, CH3), 1.36-1.43 (m, IH, pip-H), 1.56-1.83 (m, 4H, pip-H, NCH2CH2), 1.94-2.06 (m, 2H, OCH2CH, pip-Η), 2.30-2.49 (m, 10Η, pip- Η, NCH2CH2O, NCH2CH2CH2N), 2.68 (m, 1Η, pip-Η), 2.79-2.88 (m, 2Η, pip-H, CH2ArF), 2.97 (dd, IH, J = 6.8, 14.3, CH2ArF), 3.56-3.74 (m, 8H, H-4, OCH2CH, CH2Ar0IBu, NCH2CH2O), 4.60 (d, 1Η, J = 15.0, CΗ2ArOiBU), 6.84-7.20 (m, 8H, Ar-H). HPLC rR = 10.2 min (method B).
Example 260 - 4-(4-Fluoroberizyl)-3-(4-isobutoxybenzyl)-8-isopropyl-l-oxa-3,8-diaza- spiror4.51decan-2-one. hydrochloride (69NLS85)
[0808] 69NLS77 (320 mg, 0.60 mmol) was N-BOC deprotected as described in the preparation of 69NLS79-II and dissolved in DMF (3 mL). Potassium carbonate (250 mg, 1.80 mmol) was added, followed by isopropylbromide (68 μL, 0.73 mmol)) and sodium iodide (110 mg, 0.73 mmol) and the mixture stirred overnight at 500C. Workup was carried out as for 69NLS79-II. The residue was purified by silica gel column chromatography, eluting with a stepwise gradient of 0-6% methanol in dichloromethane, followed by repurification of the compound by acidic ion-exchange column, affording 69NLS85 (150 mg, 53%) as a colorless oil. The compound was converted to its hydrochloride form by treatment with 2M HCl in diethylether as described above, giving the salt as a colorless powder.
[0809] R/ = 0.75 (MeOH/CH2Cl2 1 :9). LCMS m/z 469 [M+H]+. 1H NMR (CD3OD5 free amine) δ 1.00 (d, 6H, J = 6.7, CH3), 1.01 (d, 6H, J = 6 A, CH3), 1.35-1.42 (m, IH, ρip-H), 1.56-1.60 (m, IH, pip-H), 1.72-1.79 (m, IH, pip-H), 1.93-2.06 (m, 2H, OCH2CH5 pip-H), 2.44-2.51 (m, 2H, pip-H), 2.59-2.75 (m, 3H, pip-H, NCH(CH3);.), 2.82- 2.97 (m, 2H, CH2ArF), 3.56 (t, IH, J = 6.8, H-4), 3.69-3.73 (m, 3H, OCH2CH, CH2Ar0JBu), 4.61 (d, IH, J= 15.2, CH2ArOiBu), 6.83-7.19 (m, 8H, Ar-H). HPLC tR = 11.0 min (method B).
Example 261 - 4-f4-Fluorobenzyl)-3-(4-isobutoxybenzyl)-8-f2-('2-oxo-imidazolidin-l-yl)- ethvn-l-oxa-3,8-diaza-spiror4.51decan-2-one, hydrochloride ("38PH17-HCD
[0810] The title compound was obtained as a colorless solid in 29% yield from 69NLS77 (180 mg, 0.34 mmol) following the same procedure as described for 69NLS85. Toluene-4-sulfonic acid 2-(2-oxo-imidazolidin-l-yl)-ethyl ester was used as the alkylating agent.
[0811] R7 = 0.63 (MeOH/CH2Cl2 1 :9). LCMS m/z 539 [M+H]+. 1H NMR (CD3OD, free amine) δ 1.01 (d, 6H, J= 6.8, CH3), 1.34-1.42 (m, IH5 pip-H), 1.56 (m, IH, pip-H), 1.77-1.85 (m, IH, pip-H), 1.94 (m, IH, pip-H), 1.99-2.07 (m, IH5 CH(CH3)2), 2.33-2.42 (m, 2H, pip-H), 2.55 (t, 2H, J= 6.6, NCH2CH2), 2.71 (m, IH, pip-H), 2.83-2.89 (m, 2H, pip-H, CH2ArF), 2.97 (dd, IH, J = 7.0, 14.2, CH2ArF), 3.25 (t, 2H5 J = 6.6, NCH2CH2), 2.89-3.36, 3.42-3.46 (2m, 4Η, CONCH2), 3.56 (t, IH, J = 7.0, H-4), 3.71 (d, 2H, J = 6.4, OCH2CH), 3.70 (d, IH, J = 15.0, CH2Ar0JBu), 4.60 (d, IH, J = 15.0, CH2ArOiBu), 6.84-7.20 (m, 8H, Ar-H). HPLC fR = 10.0 min (method B).
Example 262 - 4-(4-Fluorobenzyl>3-(4-isobutoxybenzvD-l-oxa-3.8-diaza- spiror4.51decan-2-one. hydrochloride (69NLS8n
[0812] 69NLS77 (300 mg, 0.57 mmol) was N-BOC deprotected as described in the preparation of 69NLS79-II. The residue was purified by silica gel column chromatography, eluting with a stepwise gradient of 0-6% methanol in dichloromethane, and purification on an acidic ion-exchange SPE cartridge, afforded 69NLS81 (127 mg, 52%) as a colorless oil. Formation of the hydrochloride salt was carried out as before for 69NLS75 giving the title compound as a colorless solid.
[0813] R/ = 0.29 (MeOH/CH2Cl2 1:9). LCMS m/z All [M+H]+. 1H NMR (CD3OD, free amine) δ 1.00 (d, 6H, J= 6.8, CH3), 1.25-1.33 (m, IH, pip-H), 1.52 (m, IH, pip-H), 1.65-1.73 (m, IH, pip-H), 1.91 (m, IH, pip-H), 1.98-2.06 (m, IH, CH(CH3);,), 2.73-2.97 (m, 6H, pip-H, CH2ArF), 3.53 (t, IH, J = 7.0, H-4), 3.68-3.71 (m, 3H, OCH2CH, CH2ArOiBu), 4.59 (d, IH, J = 15.2, CH2ArOiBu), 6.83-7.19 (m, 8H, Ar-H). HPLC tR = 9.7 min (method B).
Example 263 - 8-Cvclopropylmethyl-4-(4-fluorobenzyl')-3-(4-isobutoxybenzyl)-l-oxa- 3.8-diaza-spiror4.51decan-2-one, hydrochloride (38PH16-HC1)
[0814] The title compound was obtained as a colorless solid in 10% yield from 69NLS77 (180 mg, 0.34 mmol) following the same procedure as described for 69NLS85.
[0815] R/ = 0.64 (MeOH/CH2Cl2 1:9). LCMS m/z 481 [M+H]+. 1H NMR (CD3OD, free amine) δ 0.25, 0.62 (2m, 4H, CH(CH2)2), 0.92-0.99 (m, 1Η, CH(CΗ2)2), 1.03 (d, 6H, J= 6.6, CH3), 1.52-1.59 (m, IH, pip-H), 1.71 (m, IH, pip-H), 1.89-2.15 (m, 3H, pip-H, CH(CH3)2), 2.60 (d, 2H, J= 6.8, NCH2), 2.73-2.80 (m, 2H, pip-H), 2.90 (dd, IH, J = 7.2, 14.4, CH2ArF), 3.03 (dd, IH, J= 7.0, 14.4, CH2ArF), 3.25-3.13 (m, 2H, pip- H), 3.66 (t, IH, J = 7.2, NCH), 3.73 (d, IH, J = 6.4, OCH2CH), 3.76 (d, IH, J = 15.0, CH2ArOiBu), 4.63 (d, IH, J= 15.0, CH2AroiBu), 6.85-7.23 (m, 8H, Ar-H). HPLC /R = 11.4 min (method B).
Example 264 - 4-(4-Fluorobenzyl)-3-f4-isobutoxyben:gyl)-8-ethyl-l-oxa-3.8-diaza- spiror4.51decan-2-one. hydrochloride f38-PH20^
[0816] 69NLS77 (190 mg, 0.45 mmol) was N-BOC deprotected as described in the preparation of 69NLS79-II and dissolved in DMF (3 mL). Potassium carbonate (250 mg, 1.80 mmol) was added, followed by ethylbromide (50 μL, 0.45 mmol)) and the mixture stirred overnight at rt. Workup was carried out as for 69NLS79-II. The residue was purified by silica gel column chromatography, eluting with a stepwise gradient of 0- 6% methanol in dichloromethane, followed by repurification of the compound by acidic ion-exchange SPE cartridge, to give 38-PH20 (77 mg, 38%) as a colorless oil. The compound was converted to its hydrochloride form by treatment with 2M HCl in diethylether as described above, giving the salt as a colorless powder.
[0817] R/ = 0.52 (MeOH/CH2Cl2 1:9). LCMS m/z 455 [M+H]+. 1H NMR (CDCl3, salt) S 0.96 (d, 6H, J = 6.6, CH3), 1.39-1.55 (m, 6H, NCH2CH3, pip-H), 1.98- 2.04 (m, 2H, pip-H, GH(CH3)2), 2.75-2.86 (m, 2H, CH2ArF), 2.98 (m, 4H, NCH2CH3, pip- H), 3.19 (d, IH, J= 14.6, CH2ArOiBu), 3.25-3.50 (m, 3H, pip-H, H-4), 3.62 (d, 2H, J= 6.3, OCH2CH), 4.60 (d, IH, J = 14.6, CH2ArOiBu), 6.70-7.19 (m, 8H, Ar-H). HPLC *R = 4.7 min (method A).
Example 265 - 3-(4-Difluoromethoxyben2ΛiV4-(4-fluorobenzyO-8-methyl-l-oxa-3,8- diaza-spiro|4.5]decan-2-one, hydrochloride (84AF8-30)
[0818] The title compound was obtained as a colorless" solid in 25% yield from 69NLS44 (85 mg, 0.30 mmol) following the same procedure as described for 69NLS75.
[0819] R/ = 0.44 (MeOH/CH2Cl2 6:94). LCMS m/z 434 [M+H]+. 1H NMR (CDCl3, free amine) δ 1.42-1.50 (m, IH, pip-H),. 1.58-1.63 (m, IH, pip-H), 1.76-1.83 (m, IH, pip-H), 1.90-1.95 (m, IH, pip-H), 2.28 (m, 3H, NCH3), 2.37-2.45 (m, 2H, pip-H), 2.60 (m, IH, pip-H), 2.72 (m, IH, pip-H), 2.80 (dd, IH, J=6.8, 14.4, CH2ArF), 2.87 (dd, IH, j=7.6, 14.4, CH2ArF), 3.45 (t, IH, J = 6.8, NCH), 3.73 (d, IH, J= 15.2, CH2ArOiBu), 4.71 (d, IH, J= 15.2, CH2ArOiBu), 6.50 (t, IH, JC.F = 73.6, CF2H), 6.96-7.06 (m, 8H, Ar- H). HPLC tø. = 2.7 min (method A).
[0820] 8-Methyl-4-(4-methylbenzyl)-3-(4-trifluoromethoxybenzyl)- 1 -oxa-3,8- diaza-spiro[4.5]decan-2-one, hydrochloride (69NLS21)
Example 266 - Preparation of 4-ri-Carboxy-2-(4-methvIphenyl)-ethyll-4-hvdroxy-l- methyl-piperidine (69NLS13\
[0821] The title compound was obtained in analogy with the procedure described for 69NLS42 starting with 3-(4-methylphenyl) propionic acid (0.70 g, 4.26 mmol).
[0822] R/= 0.14 (MeOH/CH2Cl2 1 :9).
Example 267 - Preparation of 4-f4-MethylbenzylV8-rnethyl-l-oxa-3.8-diaza- spiror4.51decan-2-one f69NLS15). [0823] The title compound was obtained from 69NLS13 in analogy with the procedure described for 69NLS44.
[0824] R/= 0.5 (MeOH/CH2Cl2 1 :9). LCMS m/z 275 [M+H]+. HPLC tR = 3.8 min (method B).
Example 268 - Preparation of 8-Methyl-4-f4-methvIbenzv0-3-("4- trifluoromethoxybenzyl')-l-oxa-3,8-diaza-spiror4.51decan-2-one, hydrochloride
(69NLS2R
[0825] The title compound was obtained in 24% overall yield by alkylation of 69NLS15 with p-trifluoromethoxy benzylbromide, as described in the procedure for 69NLS75. Conversion of the free amine into the hydrochloride salt was performed as above, giving the title compound as a colorless solid.
[0826] Ry = 0.26 (MeOH/CH2Cl2 1:9). LCMS m/z 449 [M+H]+. 1H NMR (CD3OD, free amine) δ 1.46-1.53 (m, IH, pip-H), 1.64 (m, IH, pip-H), 1.81-1.89 (m, IH, pip-H), 1.97 (m, IH, pip-H), 2.24, 2.30 (2s, 6H, Ar-CH3, NCH3), 2.30-2.38 (m, 2H, pip- H), 2.63 (m, IH, pip-H), 2.74 (m, IH, pip-H), 2.85 (dd, IH, J= 6.1, 14.2, CH2ArMe), 2.92 (dd, IH, J = 7.4, 14.2, CH2ArMe), 3.67 (t, IH, J = 7.0, H-4), 3.90 (d, IH, J = 15.2, CH2ArOcF3), 4.59 (d, IH, J= 15.2, CH2Ar0CF3), 7.03-7.21 (m, 8H, Ar-H). HPLC tø = 10.9 min (method B).
Example 269 - 3-(4-Methoxybenzyl)-8-methyl-4-(4-methylbenzyl')-l-oxa-3,8-diaza- spirof4.51decan-2-one. hydrochloride r69NLS35^
[0827] The title compound was obtained in 20% yield in analogy with the procedure described for 69NLS75 by alkylation of 69NLS15 with p-methoxy benzylchloride. In this case however, the reaction was performed at reflux for 3h. Conversion of the free amine into the hydrochloride salt was performed as above, gave the title compound as a colorless solid.
[0828] Rf = 0.45 (MeOH/CH2Cl2 1:9). LCMS m/z 395 [M+H]+. 1H NMR (CD3OD, free amine) δ 1.33-1.41 (m, IH, pip-H), 1.51-1.56 (m, IH, pip-H), 1.77-1.85 (m, IH, pip-H), \.92-1.91 (m, IH, pip-H), 2.22 and 2.30 (2s, 6H, Ar-CH3, NCH3), 2.27-2.36 (m, 2H, pip-H), 2.56 (m, IH, pip-H), 2.70 (m, IH, pip-H), 2.81 (dd, IH, J = 7.2, 14.2, CH2ArMe), 2.94 (dd, IH, J= 7.0, 14.2, CH2ArMe), 3.56 (t, IH, J= 7.0,.H-4), 3.72 (d, IH, J = 15.2, CH2Ar0Me), 3.76 (s, 3H5 OCH3), 4.59 (d, IH, J= 15.2, CH2Ar0Me), 6.84-7.13 (m, 8H, Ar-H). HPLC tR = 6.2 min (method A).
Example 270 - (4R)- and (4S>-4-(4-fluoroben2ylV3-C4-isobutoχybenzyl')-8-methyl-l-oxa- 3.8-diaza-spiror4.51decan-2-one. hydrochloride (78NLS59 and 78NLS62)
[0829] The resolution of the racemate 69NLS44 was achieved by transient introduction of a camphanoyl chiral auxiliary. The resulting two diastereomers were separated by fractional crystallization, the camphanic acid substituent removed and the resulting spiropiperidines alkylated with the appropriate benzylbromide derivative.
Example 271 - Preparation of 3-[(-VCamphanoyl)]-4-f4-fluorobenzyiy8-methyl-l-oxa- 3,8-diaza-spiro[~4.51decan-2-one and separation of the two diastereomers (78NLS52-crys and 78NLS52-fϊn.
[0830] H-Butyllithium (2.7 M in heptane, 0.53 mL, 1.42 mmol) was added to a cooled solution of 69NLS44 (360 mg, 1.29 mmol) in THF (10 mL) at -780C, and the mixture stirred for 30 min. A solution of (-)-camphanic acid chloride (307 mg, 1.42 mrnol) in THF (2 mL) was added dropwise at -780C, the solution stirred for 15 min at - 780C and then 3 h at rt. Saturated ammonium chloride solution (5 mL) was added and the mixture extracted three times with ethyl acetate. The combined organic extracts were washed with sat. NaHCθ3 and brine, dried over Na2SO4, filtered and evaporated. The residue was crystallized from ethyl acetate/«-heptane at rt. The light-yellow crystals 78NLS52-crys (155 mg, 26%, >98% de, determined by 1H NMR) were filtered off, the mother liquor concentrated and purified by silica gel column chromatography, eluting with a stepwise gradient of 0-2% methanol in dichloromethane. The second diastereomer is accumulated in the head fractions, which were evaporated and the residue recrystallized as before. Evaporation of the mother liquor afforded 78NLS52-fil (28 mg, 5%, 98% de determined by 1H NMR) as a colorless oil.
[0831] 78NLS52-crys (>98% de): R/= 0.40 (MeOH/CH2Cl2 1 :9). LCMS m/z 459 [M+H]+. 1H NMR (CDCl3) δ 0.87, 1.09 and 1.15 (3s, 3x3H, CH3), 1.45-1.53 (m, IH, pip-H), 1.70-1.80 (m, 3H, pip-H, camph-H), 1.83-1.92 (m, IH, camph-H), 1.97-2.02 (m, IH, pip-H), 2.13-2.20 (m, IH, camph-H), 2.24 (s, 3H, NCH3), 2.29-2.38 (m, 2H, ρiρ-H), 2.55-2.61 (m, 2H, pip-H, camph-H), 2.66 (m, IH, pip-H), 2.80 (dd, IH, J = 9.0, 14.2, CH2Ar), 3.06 (dd, IH, J = 4.9, 14.2, CH2Ar), 4.59 (dd, IH5 J = 4.9, 9.0, H-4), 6.98-7.25 (m, 4H3 Ar-H). HPLC tR = 8.1 min (method B).
[0832] 78NLS52-fil (98% de): R/= 0.40 (MeOH/CH2Cl2 1 :9). LCMS m/z 459 [M+H]+. 1H NMR (CDCl3) δ 0.98, 1.09 and 1.13 (3s, 3χ3H, CH3), 1.59-1.70 (m, 2H, pip- H), 1.73-1.80 (m, IH, camph-H), 1.84-1.91 (m, IH, camph-H), 1.97 (m, 2H, pip-H), 2.08- 2.15 (m, IH, camph-H), 2.25 (s, 3H, NCH3), 2.24-2.37 (m, 2H, pip-H), 2.50-2.60 (m, 2H, pip-H), 2.94-3.01 (m, 2H, CH2Ar, pip-H), 3.12 (dd, IH, J= 4.1, 14.2, CH2Ar)3 4.32 (dd, IH, J= 4.1, 9.0, H-4), 6.95-7.24 (m, 4H, Ar-H). HPLC tR = 8.7 min (method B).
Example 272 - Preparation of (4R)- and ("4S)-4-(4-fluorobenzyl)-8-methyl-l-oxa-3,8- diaza-spiror4.51decan-2-one (78NLS57 and 78NLS61).
[0833] Lithium hydroxide monohydrate (20 mg, 0.47 mmol) was added to a solution of 78NLS52-crys (100 mg, 0.22 mmol) in THF/water (4 mL, 3:1) at O0C. After stirring for 1 h at 00C, sat. aq. NaHCO3 was added (3 mL), the solution extracted three times with diethylether and the combined organic extracts dried over Na2SCu, filtered and evaporated. The crude compound 78NLS57 (60 mg) was used without further purification. Tn the same way, treatment of 78NLS52-fil (28 mg, 61 μmol) with lithiumhydroxide afforded 78NLS61. Λ
Example 273 - Preparation of (4R)- and (4S)-4-("4-fluorobenzylV3-(4-isobutoxybenzylV8- methyl-l-oxa-3.8-diaza-spiroC4.51decan-2-one. hydrochloride (78NLS59 and 78NLS62V
[0834] Alkylation of 78NLS57 (ca. 0.22 mmol) following the method described for 69NLS75 gave the enantiomer 78NLS59 (45 mg, 48%, over two steps). In the same way, alkylation of the second enantiomer 78NLS61 (ca. 61 μmol) gave 78NLS62 (7 mg, 25%, over two steps). Spectrochemical data for both compounds were identical with those determined for 69NLS75. The enantiomeric excess (ee) was determined to be 98% for 78NLS59 and 93% for 78NLS62 using chiral HPLC analysis (Chiracel OD-H column, 4.6 x 250 mm; hexane/I-PrOH/DEA 95:5:0.2; 0.5 mL/min; tR 20.8 and 23.6 min for 78NLS59 and 78NLS62 respectively).
[0835] Subsequent X-ray diffraction analysis of the intermediate 78NLS52- crys allowed the assignment of the ^-configuration to the early eluting enantiomer (78NLS59, Formula II). Biological evaluation in vitro demonstrated that the (S)- enantiomer is the eutomer at the 5-HT2A receptor (See Table 6).
Figure imgf000226_0001
Example 274 - [2-(4-Fluorophenyl)ethyl]triphenylphosphonium bromide C78NLS66)
[08361 4-Fluorophenylethyl bromide (700 mg, 3.44 mmol) was dissolved in toluene (4 mL), triphenylphosphine (904 mg, 3.44 mmol) added and the solution heated for 10 min in a sealed flask at 2000C under microwave heating. After cooling to rt, the solvent was decanted and the title compound was obtained quantitatively as the remaining glassy solid, which was used without further purification.
[0837] R7= 0.70 (MeOH/CH2Cl2 1 :9). 1H NMR (CDCl3) δ 2.99-3.06 (m, 2H,
PCH2CH2), 4.16-4.23 (m, 2H, PCH2), 6.86-7.35 (m, 4H, ArF-H), 7.67-7.90 (m, 15H,
PPh3).
Example 275 - 4-[2-(4-Fluorophenyl')ethylidenel-piperidine-l-carboxylic acid benzyl ester C103NLS05)
[0838] To a suspension of phosphonium bromide 78NLS66 (4.69 g, 10.1 mmol) in THF (100 mL) H-butyllithium (1.6 M in hexane, 6.3 mL, 10.1 mmol) was added at O0C, giving a deep red solution. After stirring for 1 h at rt, a solution of benzyl 4-oxo-l- piperidinecarboxylate (2.24 g, 9.6 mmol) in THF (10 mL) was added dropwise over 30 min. The mixture was stirred at rt for 2Oh, then water was added and the mixture extracted three times with diethylether. The combined organic layers were dried over Na2SOzJ, filtered and concentrated in vacuo. Purification of the residue by silica gel column chromatography, eluting with a stepwise gradient of 0-10% ethyl acetate in M-heptane, afforded the title compound (0.56 g, 17%) as a colorless oil.
[0839] R/ = 0.63 (ethyl acetate/w-heptane 2:3). LCMS m/z 362 [M+Na]+, 340 [M+H]+. 1H NMR (CDCl3) δ 2.24 (m, 2H, pip-H), 2.35 (m, 2H5 pip-H), 3.37 (d, 2H, J = 7.2, CH2ArF), 3.55 (m, 4H, pip-H), 5.16 (s, 2H, OCH2), 5.43 (t, IH, J= 7.2, CH-), 6.96- 7.40 (m, 9H, Ar-H). HPLC tR = 10.7 min (method B).
Example 276 - 2-(4-FluorobenzylM-oxa-6-aza-spiro[2.51octane-6-carboxylic acid benzyl ester (103NLSO^
[0840] To a solution of the olefin 103NLS05 (79 mg, 0.23 mmol) in dichloromethane (3 mL) at O0C was added dropwise a solution of m-chloroperbenzoic acid (70% in H2O, 69 mg, 0.28 mmol). The mixture was stirred at rt for 24 h, diluted with dichloromethane and washed with 10% aq. NaHCO3 solution. The organic layer was dried over Na2SO4, filtered and evaporated to dryness. The title compound was obtained quantitatively and was used without further purification.
[0841] R/ = 0.53 (ethyl acetate/n-heptane 2:3). LCMS m/z 356 [M+H]+. 1H NMR (CDCl3) δ 1.39 (m, IH, pip-H), 1.66 (m, IH5 pip-H), 1.83-1.90 (m, 2H5 piρ-H), 2.79-2.92 (m, 2H, CH2ArF), 3.03 (m, IH, OCH)5 3.39-3.45 (m, 2H5 pip-H), 3.83-3.89 (m, 2H5 pip-H), 5.16 (S5 2H, OCH2), 6.98-7.36 (m5 9H5 Ar-H). HPLC tκ = 9.3 min (method B).
Example 277 - 4-[l-Amino-2-(4-fluorophenyl)-ethyl'|-4-hydroxypiperidine-l-carboxyIic acid benzyl ester flOSNLSΣδ^
[0842] To a solution of ammonia in methanol (7 N, 5 mL) the crude epoxide 103NLS09 (520 mg, 1.40 mmol) was added and the solution heated for 20 h in a sealed flask at 1000C. After cooling to rt, the solvent was removed and the residue purified by Ci8 reversed phase solid-phase extraction, eluting with a stepwise gradient of 0-80% methanol in dichloromethane, giving the aminoalcohol 103NLS28 (459 mg, 88%).
[0843] R/ = 0.62 (MeOH/CH2Cl2 1:9). LCMS m/z 373 [M+H]+. 1H NMR (CDCl3) δ 1.52-1.67 (m, 4H5 pip-H)5 2.37 (dd, IH, J= 11.3, 13.8, CH2ArF), 2.70 (m, 3H, NH2, OH)5 2.81 (dd, IH, J = 2.9, 11.3, CH2ArF), 2.97 (dd, IH, J = 2.9, 13.8, CHNH2), 3.20-3.26 (m, 2H5 piρ-H)5 4.03-4.08 (m, 2H, pip-H)5 5.15 (s, 2H, OCH2), 6.98-7.38 (m, 9H, Ar-H). HPLC tR = 5.5 min (method B).
Example 278 - 5-(4-FluorobenzvD-3-oxo-l-oxa-4,9-diaza-spiror5.51undecane-9- carboxylic acid benzyl ester C103NLS30O [0844] The title compound was prepared from aminoalcohol 103NLS2S (303 mg, 0.81 mmol) according to literature procedures (Clark et al., J. Med. Chem. 26:855- 861 (1983)), by acylation of the amino function with chloroacetylchloride, followed by halogen exchange with NaI and ring closure of the iodide derivative in presence of tert- butoxide. Purification of the crude product by silica gel column chromatography, eluting with a stepwise gradient of 0-100% ethyl acetate in n-heptane, afforded the title compound (64 mg, 19% overall yield) as a colorless solid.
[0845] R/ = 0.19 (ethyl acetate/n-heptane 3:2). LCMS m/z 458 [M+H]+. 1H NMR (CDCl3) δ 1.52-1.74 (m, 2H, pip-H), 1.87-1.98 (m, 2H5 piρ-H), 2.47 (dd, IH, J = 11.7, 13.3, CH2ArF), 2.81 (dd, IH5 J = 2.7, 13.3, CH2ArF), 3.04-3.22 (m, 2H, pip-H), 3.40-3.45 (m, IH, H-5), 4.05-4.20 (m, 4H, H-2, pip-H), 5.15 (s, 2H, OCH2Ph), 5.55 (s, IH, NH), 6.99-7.37 (m, 9H, Ar-H). HPLC tR = 12.4 min (method B).
Example 279 - 5-f4-FluorobenzylV4-(4-isobutoxybenzyl)-3-oxo-l-oxa-4,9-diaza- spiror5.5")undecane-9-carboxylic acid benzyl ester (103NLS33)
[0846] Alkylation of the spirocycle 103NLS30C (61 mg, 0.148 mmol) was performed as described in the preparation of 69NLS75 using 4-isobutoxybenzyl bromide 69NLS69 as the alkylating agent. After purification of the residue by silica gel column chromatography eluting with a stepwise gradient of 0-70% ethyl acetate in «-heptane, the title compound 103NLS33 (62 mg, 73%) was obtained as a colorless oil.
[0847] Rr = 0.37 (ethyl acetate/w-heptane 1: 1). LCMS m/z 575 [M+H]+. 1H NMR (CDCl3) δ 0.79-0.88 (m, IH5 pip-H), 1.02 (d, 6H3 J= 6.4, CH3), 1.32-1.37 (m, IH, pip-H), 1.69-1.81 (m, 2H, pip-H), 2.03-2.09 (m, IH, CH(CH3)2), 2.72 (d, IH, J = 14.3, CH2ArOiBu), 2.86-2.95 and 3.07-3.15 (2m, 5H, H-5,CH2ArF, pip-H), 3.70 (d, 2H5 J= 6.4, OCH2CH), 3.80-3.85 (m, 2H, pip-H), 4.16 (AB, 2H, J = 17.6, H-2), 5.06 (s, 2H5 OCH2Ph), 5.35 (d, IH9 J = 14.3, CH2ArOiBu), 6.78-7.36 (m, 13H, Ar-H). HPLC tR = 10.9 min (method B).
Example 280 - 5-("4-Fluorobenzyl)-4-(4-isobutoxybenzyl)-9-methyl-l-oxa-4,9-diaza- spiror5.5]undecan-3-one (103NLS35*)
[0848] The spirocycle 103NLS33 (62 mg, 0.107 mmol) in ethanol (5 mL) was N-CBz-deprotected by hydrogenation in presence of Pd/C (10%, 40 mg) under H2-balloon pressure. The catalyst was filtered off and the filtrate evaporated under reduced pressure to give crude 5-(4-fluorobenzyl)-4-(4-isobutoxyben2yl)-l-oxa-4,9-diaza-spiro[5.5]un- decan-3-one. The residue was dissolved in methanol (3 mL), formaldehyde (37% in water, 0.017 mL) added, followed by the addition of acetic acid (0.03 mL) and sodium cyanoborohydride (60 mg, 0.95 mmol). The solution was stirred at rt for 5 h, then water was added and the mixture basified with 2 N aq. NaOH solution. The mixture was extracted three times with dichloromethane, the combined organic layers washed with sat. ammonium chloride solution and brine, dried over Na2SO4, filtered and evaporated to dryness. The residue was purified by column chromatography on silica gel, eluting with a stepwise gradient of 0-7% methanol in dichloromethane, giving 103NLS35B (43 mg, 88% over both steps) as a colorless oil. The compound was converted to its HCl form by treatment with 2 M HCl in diethylether as described above for 69NLS75, affording the salt as a colorless powder.
[0849] R/ = 0.50 (MeOH/CH2Cl2 1 :9). LCMS m/z 455 [M-HH]+. 1H NMR (CDCl3) δ 0.94-1.06 (m, 7H, pip-H, CH3), 1.50-1.57 (m, IH, pip-H), 1.69-1.76 (m, 2H, pip-H), 2.00-2.09 (m, 2H3 pip-H, CH(CH3)2), 2.22-2.29 (m, 4H, NCH3, pip-H), 2.37-2.40 (m, IH, pip-H), 2.55 (m, IH, pip-H), 2.62 (d, IH, J= 14.3, CH2Ar01Bu), 2.87 (dd, IH, J= 7.8, 13.5, CH2ATF), 3.06-3.14 (m, 2H, H-5,CH2ArF, pip-H), 3.66 (d, 2H, J = 6.4, OCH2CH), 4.12 (AB, 2H, J= 17.6, H-2), 5.29 (d, IH, J= 14.3, CH2Ar01Bu), 6.75-7.18 (m, 8H, Ar-H). HPLC tκ = 7.5 min (method B).
Example 281 - 3-Oxo-l,2.8-triaza-spiror4.5]decane-8-carboxylic acid tert-butyl ester (84AF 15)
[0850] A solution of hydrazine monohydrate (5.28 mL, 108.8 mmol) in n- butanol (20 mL) was added dropwise to a solution of N-BOC-4-methoxycarbonyl- methylenepiperidine (1.39 g, 5.44 mmol) in n-butanol (120 mL) at rt. The mixture was stirred for 15 h at 120°C and the mixture allowed to cool to rt. The solvent was removed by evaporation under reduced pressure, the residue partitioned between ethyl acetate and water and the organic layer dried over sodium sulphate, filtered and evaporated to dryness. Purification of the residue by silica gel column chromatography, eluting with a stepwise gradient of 3-6 % methanol in dichloromethane, afforded the desired compound (0.88 g, 63 %) as white solid. [0851] Rr 0.34 (MeOH/CH2Cl2 6:94). LCMS m/z 200 [M+H-(t-Bu)]+, 156 [M-FH-BOC]+. 1H NMR (CDCl3) δ 1.45 (s, 9H, CH3), 1.61-1.74 (m, 4H, pip-H), 2.34 (s, 2H3 H-4), 3.38-3.52 (m, 4H, pip-H), 4.01 (s, 1 H, NH)5 7.15 (s, IH, NH).
Example 282 - l-(4-Fluoroben2ylV3-oxo-l,2,8-triaza-spiror4.51decane-8-carboxylic acid tert-butyl ester (r84AF84-191
[0852] 4-Fluorobenzylbromide (0.37 mL, 2.97 mmol) was added dropwise to a solution of 84AF 15 (289 mg, 1.13 mmol) in dry DMF (50 mL) at rt. The mixture was stirred at rt for 6 days under argon atmosphere. The solvent was removed by evaporation under reduced pressure and the residue partitioned between chloroform and water. The organic layer was dried over sodium sulphate, filtered and concentrated in vacuo. Purification of the residue by column chromatography on silica gel, eluting with dichloromethane, afforded the desired compound (160 mg, 39 %).
[0853] R/ = 0.53 (MeOHZCH2Cl2 6:94). LCMS m/z 308 [M+H-BOC]+, 364. [M+Hf. 1H NMR (CDCl3) δ 1.47 (s, 9H5 CH3), 1.71-1.76 (m, 2H5 pip-H)5 1.87-1.93 (m5 2H, pip-H), 2.45 (s. 2H5 H-4), 3.31-3.38 (m, 2H, pip-H), 3.64-3.70 (m, 2H, pip-H), 3.80 (S5 2H, CH2ArF), 6.62 (s, IH, NH), 7.01-7.30 (m, 4H, Ar-H).
Example 283 - l-(4-FluoroberιzylV2-(4-isobutoxybenzviy3-oxo-1.2.8-triaza- spiro("4.51decane-8-carboxylic acid tert-butyl ester Q18AF94-23)
[0854] NaH (24 mg, 0.50 mmol) was added slowly to a solution of 84AF84-19 (0.149 g, 0.41 mmol) in dry DMF (5 mL) and the mixture stirred at rt for 30 min. A solution of 4-isobutoxybenzyl bromide 69NLS69 (117 mg, 0.48 mmol) in dry DMF (1 mL) was added dropwise to the mixture. After 1 h stirring at rt the mixture was partitioned between ethyl acetate and water. The organic layer was dried over sodium sulphate, filtered and concentrated in vacuo. Purification of the residue by column chromatography on silica gel, eluting with a stepwise gradient of 3-6 % methanol in dichloromethane, followed by preparative reversed phase HPLC (Ci s) afforded the desired compound (109 mg, 50 %).
[0855] R7- 0.51 (MeOH/CH2Cl2 1:99). LCMS m/z 526 [M+H]+, 470 [M+H- (t-Bu)]+. 1H NMR (CDCl3) δ 1.00 (d,.6H, J = 6.8, CH(CT^)2), 1.35 (m, 4H, pip-H), 1.40 (s, 9H. O(CH3)3)5 2.06 (m, IH, CH(CH3)2), 2.41 (s, 2H5 H-4). 3.08-3.15 (m, 2H, pip-H), 3.22-3.29 (m, 2H, pip-H), 3.67 (d, 2H, J= 6.4, OCH2), 3.92 (m, 4H, CH2ArF1 CH2Ar01Bu), 6.77 -7.37 (m, 8H, Ar-H).
Example 284 - l-(4-FluorobenzylV2-(4-isobutoxybenzylVl,2.8-triaza-spiror4.51decane-3- one (84AF99-24)
[0856] 118AF94-23 (100 mg, 0.21 mmol) was N-BOC deprotected as described in the preparation of 69NLS79-II to give the title compound, which was used without further purification.
[0857] R/ = 0.40 (MeOH/CH2Cl2 1 :9). LCMS m/z 426 [M+H]+. 1H NMR (CDCl3) δ 0.99 (d, 6H, J = 6.4, CH(CHj)2), 1.35 (m, 4H, pip-H), 1.49 (s, IH, NH), 2.00- 2.05 (m. IH, CH(CH3)2), 2.42 (s, 2H, H-4), 2.50 (m, 2H, pip-H), 2.76 (m, 2H, pip-H), 3.66 (d, 2H, J= 6.4, OCH2), 3.92 (s, 2H, CH2Ar), 3.95 (s, 2H, CH2Ar), 6.75 -7.34 (m, 8H, Ar-H).
Example 285 - l-(4-FIuorobenzyl)-2-(4-isobutoxybenzyl)-8-methyl-1.2,8-triaza- spiror4.51decane-3-one. hydrochloride (84AF 100-25)
[0858] The desired spirocycle was obtained from 84AF99-24 (60 mg, 0.14 mmol) following the same procedure described for the preparation of 103NLS35. Formation of the hydrochloride salt was carried out as for 69NLS75 giving the title compound (39 mg, 63%) as a colorless solid.
[0859] R/ = 0.57 (MeOH/CH2Cl2 1 :9). LCMS m/z 440 [M+H]+. 1H NMR (CDCl3) δ 0.94 (d, 6H3 J = 6.4, CH(CHu)2), 1.38-1.68 (m, 4Η, pip-H), 1.96-2.02 (m, IH, CH(CH3)2)5 2.23-2.46 (m, 9H, pip-H, NCH3, H-4), 3.61 (d, 2H, J= 6.4, OCH2), 3.85 (m, 4H, CH2Ar01Bu, CH2ArF), 6.70-7.28 (m, 8H, Ar-H).
Example 286 - 8-(2-ri.31-Dioxan-2-yl-ethyl)-4-f4-fluorobenzyl)-3-f4-isobutoχy-bengyl)- l-oxa-3,8-diaza-spiror4.51decane-3-one, oxalate (118AF02-74)
[0860] 69NLS77 was N-BOC deprotected as described in the preparation of 69NLS79-II and the resulting compound (172 mg, 0.40 mmol) alkylated with 2-(2- bromoethyl)-l,3-dioxane following the same procedure as for the preparation of 69NLS85. Conversion of the compound (139 mg, 64%) into its oxalate salt form gave the title compound as a colorless solid. [0861] R/ = 0.33 (MeOH/CH2Cl2 1:24). LCMS m/z 541 [M+H]+. 1H NMR (CDCl3) δ 1.00 (d, 6H5 J = 6.8, CH(CHOi), 1-23-1.36 (m, 2H, pip-H, dioxane-H), 1.54- 1.66 (m, IH5 pip-H), 1.67-1.74 (m, 3H, CH2CH2N, pip-H), 1.86-1.90 (m, IH, pip-H), 1.97-2.15 (m, 2H, dioxane-H, CH(CH3)2), 2.31-2.43 (m, 4H, CH2CH2N5 pip-Η), 2.55 (ra5 1Η, pip-Η)5 2.69 (m, IH, pip-H), 2.71-2.89 (m, 2H5 CH2ArF), 3.36 (t, IH5 J = 6.8, H-4), 3.56 (d, IH, J= 15.2, CH2ArOiBu), 3.67-3.74 (m, 4H, OCH2CH(CH3)2, dioxane-H), 4.03- 4.07 (m, 2H, dioxane-H), 4.51 (t, IH5 J= 5.2, OCHO)5 4.72 (d, IH, J= 15.2, CH2ArOiBu), 6.79 -7.04 (m, 8H, Ar-H).
Example 287 - 4-f4-Fluorobenzyiy3-f4-isobutoxy-benzyl)-8--f3-r(SV4-isopropyl-2-oxo- oxazolidin-3-vn-propyU-l-oxa- 3,8-diaza-spiro|"4.51decane-3-one, oxalate Cl 18AF04-75)
[0862] 69NLS77 was N-BOC deprotected as described in the preparation of 69NLS79-II and the resulting compound (172 mg, 0.40 mmol) alkylated with (4S)-3-(3- chloropropyl)-4-isopropyl-2-oxazolidin-2-one in DMF (3 mL) in presence of sodium iodide (72 mg, 0.48 mmol), following the same procedure as for the preparation of 69NLS85. Conversion of the compound (130 mg, 55%) into its oxalate salt form gave the title compound as a colorless solid.
[0863] R/ = 0.29 (MeOH/CH2Cl2 1:19). LCMS m/z 596 [M+H]+. 1H NMR (CDCl3) δ 0.84- 0.88 (m, 6H, CH3iPr)5 1.01 (d5 6H5 J = 6.8, CH3iBu), 1.24-1.34 (m, IH, pip-H), 1.55-1.74 (m, 4H, pip-H, CH2chain), 1.87-1.93 (m, IH, pip-H), 1.97-2.13 (m, 2H, CHoiBu, CHiPr), 2.27-2.40 (m, 4H, pip-H, NCH2chain), 2.56 (m, IH, pip-H), 2.69 (m, IH, pip-H), 2.75-2.80 (m5 IH, CH2ArF), 2.87-2.98 (m, 2H, CH2ArF, NCH2chain), 3.37 (t, IH, J = 6.8, H-4), 3.48-3.55 (m, IH, NCH2ohain)> 3.58-3.64 (m, IH5 CH2AroiBu), 3.65-3.71 (m, 3H, OCH2CH(CHa)2, CHisox), 4.00-4.04 (m, IH, OCHiS0X), 4.1 1-4.16 (m, IH, OCHisox), 4.75 (d, IH5 J= 15.2, CH2Ar0IBu), 6.80-7.03 (m, 8H5 Ar-H).
Pharmacological Data
Example 288 - Receptor Selection and Amplification (R-SAT) Assays.
[0864] The functional receptor assay, Receptor Selection' and Amplification Technology (R-SAT)5 was used (with minor modifications from that previously described US 5,707,798, which is incorporated herein by reference in its entirety) to screen compounds for efficacy at the 5-HT2A receptor. The RSAT assay was conducted as described herein. The results obtained for several compounds of the invention are presented in Table 6, below.
Table 6. Efficiency and pIC50 of Compounds at the 5-HT2A Receptor Compared to Ritanserin.
Figure imgf000233_0001
Example 289 - Selectivity Profile for 4-(4-Fluorobenzyl>3-(4-isobutoxybenzyl>8-methyl- 1 -oxa-3,8-diaza-spiror4.51decan-2-one (69NLS75^
[0865] The R-SAT assay (described above in example 289) was used to investigate the selectivity of 4-(4-Fluoro-benzyl)-3-(4-isobutoxy-benzyl)-8-methyl-l-oxa- 3,8-diaza-spiro[4.5]decan-2-one. The results from a broad profiling of this compound at a variety of receptors are reported in Table 7 below. NR means No Response, i.e. the compound investigated showed no effect at the receptor studied.
Table 7. Selectivity of 4-(4-Fluorobenzyl)-3-(4-isobutoxybenzyl)-8-rnethyl-l-oxa-3,8- diaza-spiro[4.5]decan-2-one.
Figure imgf000234_0001
Example 290 - Preparation of N-f4-fluorobenzylVN-α-memylpiperidin-4-ylVN'-(4-f2- methylpropyloxy^phenylmethvDcarbamide
N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl)carbamide a) Preparation of
Figure imgf000235_0001
[0866] Triacetoxy borohydride (6.5 kg) was added over 1.5 h to a solution of N-methylpiperid-4-one (3.17 kg) and 4-fluorobenzylamine (3.50 kg) in methanol (30 L) maintaining the temperature under 27 0C. The reaction mixture was stirred for 15 h at 22 0C. The residual amine was checked by gel chromatography (4-fluorobenzylamine: < 5%). A solution of 30% sodium hydroxide (12.1 kg) in water (13.6 kg) was added in 75 minutes (min) maintaining the temperature under 20 0C. Methanol was distilled off to a residual volume of 26 litres. Ethyl acetate was added (26 L), the solution was stirred for 15 min, the phases were decanted over 15 min and the lower aqueous phase was discarded. Ethyl acetate was distilled under reduced pressure from the organic phase at 73-127 0C. At this stage the residue was mixed with a second crude batch prepared according to this method. The combined products were then distilled at 139-140 0C / 20 mbar to yield 11.2 kg product (> 82%).
b) Preparation of
Figure imgf000235_0002
[0867] 4-Hydroxybenzaldehyde (4.0 kg) and ethanol (20 L) were added to a solution of isobutyl bromide (9.0 kg) in ethanol (15 L). Potassium carbonate (13.6 kg) was added and the suspension was refluxed (74-78 0C) for 5 days. The residual 4- hydroxybenzaldehyde was checked by HPLC (< 10%). The suspension was cooled to 20 0C and used in the next step. c) Preparation of
Figure imgf000236_0001
[0868] Hydroxylamine (50% in water, 8.7 kg) was added to the product from previous step b) (174 L, 176 kg) and ethanol (54 L). The suspension was refluxed (77 0C) for 3 h. Unreacted residual was checked by HPLC (< 5%). The suspension was cooled to 30 0C, filtered and the filter was washed with ethanol (54 L). The solution was concentrated by distillation under reduced pressure at 30 0C to a residual volume of 67 litters. The solution was cooled to 25 0C and water (110 L) was added. The suspension was concentrated by distillation under reduced pressure at 30 0C to a residual volume of 102 litters. Petrol ether (60-90 fraction, 96 L) was added and the mixture was heated to reflux (70 0C). The solution was cooled to 40 0C and crystallization was initiated by seeding. The suspension was cooled to 5 0C and stirred for 4h. The product was centrifuged and the cake was washed with petrol ether (60-90 fraction, 32 L). The wet cake was dried at about 400C to yield 16kg product (63%) .
d) Preparation of
Figure imgf000236_0002
[0869] The product from previous step c) (15.7 kg) was dissolved in ethanol (123 L). Acetic acid (8.2 kg) and palladium on charcoal 5% wet (1.1 kg) were added. The oxime was hydrogenated at 22 0C and 1.5 bar for 4h. Consumption of oxime was checked by HPLC. The catalyst was filtered and the solvent was distilled under reduced pressure at 36 0C to a final volume of 31 L. Ethyl acetate (63 L) was added and the mixture was heated to reflux (75 0C) until dissolution. The solution was cooled to 45 0C and the crystallization was initiated by seeding. The suspension was cooled to 6-10 0C and stirred for 2.5h. The product was centrifuged and the cake was washed with 2 portions of ethyl acetate (2 x 0.8 L). The wet cake was dried at a temperature of about 40 0C to yield 8 kg (41%). e) Preparation of
Figure imgf000237_0001
[0870] Aqueous sodium hydroxide (30%, 5.0 kg) was added to a suspension of the product from previous step d) (7.9 kg) in heptane (41 L). The solution was heated to 47 0C, stirred for 15 min and decanted over 15 min. The pH was checked (pH>12) and the aqueous phase was separated. The solvent was removed by distillation under reduced pressure at 47-65 0C. Heptane was added (15 L) and then removed by distillation under reduced pressure at 58-65 0C. Heptane was added (7 L), the solution was filtered, and the filter was washed with heptane (7 L). The solvent was removed by distillation under reduced pressure at 28-60 0C. Tetrahydrofuran (THF, 107 L) and triethylamine (TEA, 6.8 kg) were added and the temperature was fixed at 22 0C. In another reactor, phosgene (5.0 kg) was introduced in tetrahydrofuran (88 L) previously cooled to -3 0C. The THF and TEA solution was added to the solution of phosgene in 3h 50 min, maintaining the temperature at -3 0C. The reactor was washed with tetrahydrofuran (22 L). The mixture was stirred for 45 min at 20 0C and then for 90 min at reflux (65 0C). The solvent was distilled under reduced pressure at 25-30 0C to a residual volume of 149 L. The absence of phosgene was controlled. At this stage, phosgene was still present and the suspension was degassed by bubbling nitrogen through it. After this operation, the level of phosgene above the solution was below 0.075 ppm. The suspension was filtered and washed with tetrahydrofuran (30 L). The solvent was distilled under reduced pressure at 20-25 0C to a residual volume of 40 L. Tetrahydrofuran (51 L) was added and the solvent was distilled under reduced pressure at 20-25 0C to a residual volume of 40 L. The final volume was adjusted to about 52 litters by addition of tetrahydrofuran (11 L). The solution was analysed and used in the next step.
f) Preparation of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy) phenylmethyl) carbamide
Figure imgf000238_0001
[0871] The product from previous step e) (51 L) was added in 1 h to a solution of the product from step a) (7.3 kg) in tetrahydrofuran (132 L) at 17 0C. The line was washed with tetrahydrofuran (12 L) and the mixture was stirred for 15h. Residual product from the first step was checked by HPLC. The solvent was removed by distillation under reduced pressure at 20-38 0C to a residual volume of 165 L. Charcoal (Norit SXl-G5 0.7 kg) was added, the mixture was stirred for 15 min and filtered. The line was washed with tetrahydrofuran (7 L) and the solvent was removed by distillation under reduced pressure at 20-25 °C to a residual volume of 30 L. Isopropyl acetate (96 L) was added to obtain a solution of the title compound, which contains a small amount of impurities (mainly side products from the previous reactions.) Removal of the solvent from a sample yields a substantially amorphous solid.
[0872] The solution with the crude product was used for the direct preparation of the hemi -tartrate and simultaneously for the purification of the free base via the hemi- tartrate through crystallization from suitable solvents.
Example 291 — R-SAT Assay of N-(l-methylpiperidin-4-vπ-N-('4-fluorophenylmethyl)- N'-(4-f2-methylpropyloxy*)phenylmethvO carbamide
[0873] The functional receptor assay Receptor Selection and Amplification Technology (R-SAT) was used to investigate the activity of N-(I -methylpiperidin-4-yl)- N-(4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide as an inverse agonist at 5HT2A receptors. The compound exhibited high potency (pIC50 of 9.1) and high efficacy (98%) at 5HT2A receptors. Example 292 - Effects of N-α-methylpiperidin^-yiyN^-fluorophenylmethylVN'^^- methylpropyloxy'Ϊphenylmethyl) carbamide on Slow Wave Sleep and Other Sleep Parameters
[0874] A double-blind, placebo-controlled study involving 45 healthy volunteers ranging in age from 40 to 64 was conducted. The purpose of the study was to determine the effects of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide on slow wave sleep and other sleep parameters as measured by polysomnography (PSG) in older adults. The effects of N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide were measured before and after a single dose and at plasma steady state. The subjects were randomized to one of five treatment arms, including placebo and four different doses (1 mg, 2.5 mg, 5 mg, and 20 mg) of N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide. Each group was administered placebo or the specified dose of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide once-daily each morning for 14 consecutive days. All subjects underwent a two-night screening and baseline PSG evaluation. Additional PSG measurements were performed on the evening of study days 1 and 13. The subjects also completed a Continuous Performance Test (CPT) to assess the potential impact on daytime functioning. Blood samples were collected at the beginning and end of the 14-day period to determine N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'- (4-(2-methylpropyloxy)phenylmethyl) carbamide levels in plasma. Of the 45 subjects, 20 of them also underwent a positron emission tomography (PET) study to measure 5-HT2A brain receptor occupancy.
Ϊ0875] Inclusion criteria included a reported bedtime between 9 pm and midnight, a usual sleep duration. >4 hours, and habitual time in bed between 6 and 9 hours per night. Women had to be postmenopausal or previously surgically sterilized in order to participate. Exclusion criteria included a history or current diagnosis of other sleep disorders (such as restless leg syndrome, periodic leg movements with arousals, narcolepsy, rapid eye movement (REM) Behavior Disorder, circadian rhythm sleep disorder, breathing related sleep disorder, or parasomnias), smoking more than 10 cigarettes (or equivalent) per day, a history of alcohol or substance abuse or positive drug or alcohol screen, or drinking more than 5 cups of caffeine containing beverages, more than 1 liter of xanthine containing beverages, or more than 40 g of alcohol per day. Participants with an occupational history that included shift work or recent significant travel across three or more time zones within the prior two weeks or self reported napping of >45 min per day were also excluded. In addition, after a screening PSG, participants with an apnea-hypopnea index > 15 or a periodic leg movement arousal index > 15 were also excluded.
[0876] Twenty-three (23) men and 22 women were included in the study. The mean (± standard deviation) age of the participants was 51.8 ± 6.9 years (range 40 to 64). The mean BMI was 24.05 ± 2.72 kg/m2. The mean height was 168.8 ± 10 cm, and the mean weight was 68.7 ± 10.4 kg. All 45 participants completed the study and were included in data analysis.
[0877] Among the 45 participants included, 34 were non-smokers and 11 were smokers in whom the mean daily consumption was between 3.8 and 9.2 cigarettes. Eight participants drank alcohol with a mean daily consumption between 4.1 and 28.5 g. Thirty-nine of the 45 drank beverages containing caffeine with daily consumption between 1.1 and 3.5 cups. Lastly, 6 drank beverages containing xanthine with daily consumption was between 0.2 and 0.8 L. These values were not within the range for exclusion and none of these were thought to interfere with study results.
[0878] During an assessment period, participants stayed overnight in the clinic for one night from Day -1 to Day 2 and one night from Day 13 to Day 14. Participants underwent a one-night PSG evaluation on the evening of Day 1, following the first dose of treatment and a final one-night PSG evaluation on the evening of Day 13. Between Day 3 and Day 12, participants were at home.
[0879] The PSG recordings consisted of an all night recording of sleep parameters including two channels of EEG (C3 and Ol referred to contralateral mastoid), two channels of EOG (with electrode placement at the outer canthi in a superior and inferior position), and a single channel of EMG (with electrode placement in a sub-mental location). Polysomnographic recordings were performed from 23:00 to 07:00 on the following day.
[0880] Drug Dosing: A single dose of a solution of placebo or one of the four doses of N-( 1 -methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide was administered between 7:30 and 9:00 in the morning every day for 14 consecutive days. The drug was dispensed as a solution (in vials) per os (p.o.), in a double-blind fashion as determined by the randomization schedule. The content of the vials was completely consumed followed by the consumption of one vial of water.
[0881] Performance Assessment: A Continuous Performance Test (CPT; Rosvold ME, Mirsky AF, Sarason I, Bransome ER Jr., Beck LH (1956) A continuous performance test for brain damage. J Consult Psychol 20:343-350, which is incorporated herein by reference in its entirety), was used to evaluate attention and vigilance. The test was administered both during the evening and the morning following the baseline PSG on Day 1 and the evaluation PSG on Day 13. Participants were rapidly presented a numerical train with a target number as point of reference. Participants pushed a button rapidly when the target number appeared in the number list. The procedure lasted three minutes. The number of targets detected and the number of false alarms were assessed.
[0882] Safety: 3-positional vital signs, ECGs, body temperature, and respiratory rates as well as blood chemistries, hematology, and urinalysis served as safety measures. Safety measures were assessed at screening, baseline (Day -1), Day 1, Day 2, Day 13, and Day 14. Any observed or reported adverse events were recorded. Blood samples were taken on Day —1 (baseline) and on Day 14 (steady state). Two weeks after the termination of treatment administration (Day 28), an end-of-study visit was conducted, and included a clinical safety evaluation.
[0883J Plasma Levels: Blood samples were collected into Vacutainer® (dry sodium heparin, green top tube, 6 mL capacity) on Day —1 and again on Day 14 (pre- dose). The time interval between blood collection and freezing of the plasma was no longer than 45 minutes. Within 45 minutes of collection, the plasma fractions were separated by centrifugation at 3,000 rotations per minutes for 10 minutes. The plasma fractions were divided and transferred into two properly labeled storage tubes and frozen at approximately — 700C. Plasma samples were evaluted for N-(l-methylpiperidin-4-yl)- N-(4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide using a validated high performance liquid chromatography/tandem mass spectrometric (LC/MS/MS) assay.
[0884] Data Analysis: A multivariate analyses of variance (MANOVA) with 5 clusters of sleep parameters as dependent variables (sleep continuity architecture and profile parameters for the visual analysis, and NREM and REM parameters for the spectral power analysis as shown in Table 1) was conducted initially. Change from baseline to the last day of drug administration was assessed by an analysis of covariance with repeated measures (ANCOVA-RM), with the baseline variables as covariates, to detect group differences (drug at each dose versus placebo) in the PSG sleep findings. For each ANCOVA, two main effects (Treatment, 4 levels, and Study Day, 2 levels) and their interaction were analyzed. Drug to placebo contrasts were calculated as a whole (Day 1 and Day 13 combined) as well as separately for each Study Day. Change from baseline to the last day of drug administration, by group, on the results of the CPT was also analyzed. Additional pharmacodynamic analysis, including the electrographic results obtained on Study Day 1, was performed. SAS® Version 8.2 was used for non- pharmacokinetic parameters and WinNonLin V4.0 was used to compute all pharmacokinetic parameters.
Results
[0885] Compliance with the dosing regimen was reported to be high and was confirmed with the determination of drug plasma concentrations on the morning of Day 14. The mean plasma steady-state concentrations increased with increased dose and ranged from 1.6 ng/mL to 35.0 ng/mL (Table 8).
Table 8. Pharmacokinetic exposure of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide after 13 days once daily administration. Concentrations given in ng/mL (n=9/dose).
Figure imgf000242_0001
[0886] MANOVA on treatment by study day effects resulted in significant treatment effects for all clusters of sleep parameters except sleep continuity variables, suggesting that N-( 1 -methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N '-(4-(2- methylpropyloxy)phenylmethyl) carbamide significantly influenced, as a whole, most of the sleep parameters under study. A Study Day effect was evidenced only for NREM relative parameters for the spectral power density suggesting that, as a whole, NREM spectral power was influenced by time (single versus repeated treatment administration). No Study Day effect was observed for the 4 other clusters suggesting that most of the sleep EEG parameters under study were not influenced by time.
[0887] Results of the ANCOVA on the primary endpoint, slow wave sleep or stages 3 and 4 combined, showed a significant (p < 0.001) effect of Treatment and no main effect of Study Day nor Treatment by Study Day interaction, suggesting that the treatment effect was similar across the 2 study days and that the 4 treatment conditions were not differently influenced by study day. Figure 1 is a graph depicting the duration slow wave sleep observed for the various administrations (BL = baseline, Dl = Study Day I5 D13 = Study Day 13). Table 9 provides the slow sleep duration numbers. The graph shows the baseline duration as well as the absolute change from baseline. Table 10 provides the change in slow wave sleep from baseline in minutes, percentage, and statistical significance between each dose and placebo.
[0888] These results show the slow wave sleep increase seen with administration of N-(I -methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide. The data demonstrated that once-daily administration of 5 mg and 20 mg of N-(l-methyIpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide, the two highest doses used in this study, induced statistically significant increases in slow wave sleep (pO.OOl) as defined by the time spent in Stage 3 and Stage 4 sleep. These stages are commonly referred to as deep, or slow wave, sleep. This robust effect was demonstrated both acutely (on study day 1) and after chronic administration (on study day 13). The two lower doses of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'- (4-(2-methylpropyloxy)phenylmethyl) carbamide were less effective than the two higher doses, demonstrating that the sleep effects of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide were dose- related. No significant differences between study days were found, suggesting that N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide increased slow wave sleep after a single dose and that the effect was maintained with repeated administration. As depictd by the graph in Figure 2, a positive correlation was observed (r = 0.51, 95% CI = 0.22 - 0.72; r2 = 0.26, p — 0.002) between slow wave sleep duration and drug plasma level. Table 9. Effects of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide on slow wave sleep duration, number of awakenings, and wake after sleep onset duration.
Figure imgf000244_0001
SWS: slow wave sleep.
Table 10. Change in Slow Wave Sleep From Baseline in Minutes, Percentage, and Statistical Significance Between Each Dose and Placebo
Figure imgf000245_0001
[0889] Sleep Continuity Parameters: In agreement with MANOVA results showing no significant effect of treatment, ANCOVAs performed on each sleep continuity parameter taken separately were not significant for Treatment, for Study Day, or for Treatment by Study Day interaction. The sole exception was the number of awakenings after sleep onset for which a significant (p < 0.05) effect of treatment was evidenced, consistent with a shift to deeper sleep. Figure 3 is a graph depicting the number of awakenings after sleep onset. The graph shows the baseline number as well as the absolute change from baseline. This data is also presented in Table 9. Drug to placebo contrasts indicated that the 4 doses of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N' -(4-(2-methylpropyloxy)phenylmethyl) carbamide significantly decreased number of awakenings after sleep onset, with the most significant effects being observed in the 2.5 mg and 5 mg groups. The effects of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide on number of awakenings were more pronounced on Study Day 1 than on Study Day 13. A trend level of minutes awake after sleep onset (WASO), the other hallmark of sleep maintenance insomnia, was observed in the group of older healthy adults (Table 9). Other sleep continuity variables that were measured but not affected by N-( 1-methy lpiperidin-4-y I)-N- (4-fluorophenylmethyl)-N' -(4-(2-methylpropyloxy)phenylmethyl) carbamide included sleep period time, total sleep time, sleep onset latency, number of stage shifts, total time awake, early morning wake, sleep efficiency index, and microarousal index.
[0890] Sleep Architecture Parameters: In agreement with MANOVA results showing a significant Treatment effect (p < 0.001), ANCOVAs evidenced significant Treatment effects for most of the sleep architecture parameters: increased non-REM sleep duration (p < 0.05), increased non-REM sleep proportion (p < 0.01), decreased stage 2 (duration as well as proportion, p < 0.05), increased stage 3 (duration and proportion, p < 0.05) and stage 4 (duration and proportion, p < 0.001), and increased slow wave sleep proportion (stages 3 and 4 combined, p < 0.001). The direction and intensity of effects are shown in Table 11. For all sleep architecture parameters, neither significant Study Day effects nor Treatment by Study Day interactions were found. N-(l-methylpiperidin- 4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide had no effect on REM sleep duration, proportion of REM sleep, REM sleep latency, REM activity or REM density.
Table 11. Effects of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- meth l ro lox hen lmeth l carbamide on slee architecture and rofile arameters.
Figure imgf000246_0001
Figure imgf000247_0001
C: combined visits; Dl: Study Day 1; D 13: Study Day 13.
REM: rapid eye movement; NREM: non-rapid eye movement; SWS: slow wave sleep.
[0891] Sleep Profile Parameters: The MANOVA performed on sleep profile parameters showed a significant treatment effect (p < 0.001). Results of the ANCOVAs revealed that this treatment effect mostly related to slow wave sleep parameters since significant treatment effects were observed on duration of slow wave sleep on the first (p < 0.01) and second (p < 0.001) third of the night. The direction and intensity of effects are shown in Table 11. There were no other treatment effects for the other sleep profile parameters. For all ANCOVAs, neither significant Study Day effect nor a Treatment by Study Day interaction were observed.
[0892] Spectral Power Density Parameters: MANOVAs performed on spectral power density parameters evidenced a significant Treatment effect (p < 0.01) without Study Day effect during REM sleep and a significant Treatment effect (p<.001) with a significant (p < 0.001) Study Day effect during NREM sleep. For REM sleep parameters, the sole significant ANOVAs treatment effect was for Betal (p < 0.05). For non-REM sleep parameters, significant (p < 0.001) treatment effects were evidenced for slow delta, fast delta, slow wave activity, theta, spindle frequency activity and Betal (data not shown).
[0893] Performance: On the CPT, ANCOVAs indicated an absence of effect of any dose of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)ρhenylmethyl) carbamide on the detected targets and false alarm change scores, regardless evaluation session and group (p > 0.10). None of the planned comparisons were significant or approached statistical significance (p > 0.10). For the evening evaluation, a p value of 0.67 was determined for accurate detections and a p value of 0.51 was determined for false alarms. For the morning evaluation, a p value of 0.50 was determined for accurate detections and a p value of 0.87 was determined for false alarms.
Conclusion
[0894] Morning administration of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide resulted in a significant dose-related increase of slow wave sleep during the subsequent night. This effect was observed after a single administration and was sustained over time with repeated administration. Results of the clinical trial also showed that treatment with N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N ' -(4-(2- methylpropyloxy)phenylmethyl) carbamide produced trends for improvement on measures for sleep maintenance, including decreases in the number of awakenings (p=0.04) and in time awake after sleep onset (p=0.09). Importantly, in contrast to most currently marketed insomnia drugs, which are sedative, sleep-inducing agents and have the potential to impair daytime functioning, N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide did not alter latency to sleep onset and did not impair daytime functioning.
[0895] Doses of 5 mg and 20 mg and associated mean plasma levels ranging from 8.5 to 35.0 ng/mL showed robust and significant increases in different parameters directly related to slow wave sleep including slow wave sleep (stages 3 and 4 combined) duration and proportion, duration and proportion of stage 4 and the slowest components of the NREM sleep EEG activities. Increased plasma levels generally were associated with higher magnitude of effects. The highest dose of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide (20 mg) also decreased duration and proportion of wake after sleep onset. These dose dependent observations are consistent with a shift to deeper sleep. Other PSG measures such as sleep onset latency, total sleep time, sleep efficiency and REM sleep parameters were unaffected by N-(I -methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide treatment in this population.
[0896] N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide was safe and well tolerated in the study subjects and there were no serious adverse events reported. All adverse events were mild to moderate in nature and were comparable across the placebo and N-(l-methylpiperidin- 4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide - treated groups.
[0897] Although the invention has been described with reference to embodiments and examples, it should be understood that numerous and various modifications can be made without departing from the spirit of the invention. Accordingly, the invention is limited only by the following claims.

Claims

WHAT IS CLAIMED IS:
1. Use of N-(l-methylpiρeridin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide for the preparation of a medicament for treating insomnia.
2. The use of Claim 1, wherein the amount of N-(l-methylpiperidin-4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide is in the range of about 0.5 mg to about 50 mg.
3. The use of Claim 1, wherein the amount of N-(l-methylpiperidin-4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide is in the range of about 1 mg to about 40 mg.
4. The use of Claim I5 wherein the amount of N-(l-methylpiperidin-4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide is in the range of about 2.5 mg to about 30 mg.
5. The use of Claim 1, wherein the amount of N-(I -methy lpiperidin-4-y I)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide is in the range of about 5 mg to about 20 mg.
6. The use of Claim 1, wherein the N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide does not affect a sleep parameter selected from the group consisting of sleep period time, total sleep time, sleep onset latency, number of stage shifts, total time awake, early morning wake, sleep efficiency index, microarousal index, and a REM sleep parameter.
7. The use of Claim 6, wherein the REM sleep parameter is selected from the group consisting of REM sleep duration, proportion of REM sleep, REM sleep latency, REM activity and REM density.
8. The use of Claim 1, wherein the amount of N-(l-methylpiperidin~4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide results in a steady state blood plasma concentration of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the range of about 2 ng/mL to about 60 ng/mL.
9. The use of Claim 1, wherein the amount of N-(l-methyIpiperidin-4~yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide results in a steady state blood plasma concentration of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the range of about 4 ng/mL to about 50 ng/mL.
10. The use of Claim 1, wherein the amount of N-(I -methylpiperidin-4-y I)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenyLmethyl) carbamide results in a steady state blood plasma concentration of N-(l-methylpiρeridin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the range of about 6 ng/mL to about 40 ng/mL.
11. The use of Claim 1, wherein the amount of N-(I -methylpiperidin-4-y I)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide results in a steady state blood plasma concentration of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the range of about 8.5 ng/mL to about 35 ng/mL.
12. The use of Claim 1, wherein the medicament is suitable for administration once every day.
13. The use of Claim 1, wherein the medicament is provided in a first dosage form and a second dosage form of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)- N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide said first dosage form being adapted for administration at least 24 hours prior to administration of the second dosage form.
14. The use of Claim 13, wherein the first dosage form contains a higher dose of N-(I -methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide than the second dosage form.
15. The use of Claim 1, wherein the medicament is provided along with information indicating that N-(I -methylpiperidin-4-y l)-N-(4-fluorophenylmethyl)-N' -(4- (2-methylpropyloxy)phenylmethyl) carbamide ameliorates insomnia.
16. The use of Claim 15, wherein the information comprises printed matter that advises that N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide ameliorates insomnia.
17. The use of Claim 16, wherein said printed matter is a label.
18. The use of Claim 1, wherein the medicament is suitable for administration at a time other than immediately before a sleep period.
19. The use of Claim 18, wherein the medicament is suitable for administration in the morning.
20. Use of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide for the preparation of a medicament for increasing slow- wave sleep, wherein the amount of N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the medicament results in a steady state blood plasma concentration of N-(l-methylpiperidin- 4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the range of about 2.0 ng/mL to about 60.0 ng/mL.
21. The use of Claim 20, wherein the amount of N-( 1 -methylpiperidin-4-yl)-N- (4-fiuorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the medicament results in a steady state blood plasma concentration of N-(l-methylpiperidin- 4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the range of about 4.0 ng/mL to about 50.0 ng/mL.
22. The use of Claim 20, wherein the amount of N-(I -methylpiperidin-4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the medicament results in a steady state blood plasma concentration of N-(l-methylpiperidin- 4-y l)-N-(4-fluoropheny lmethyl)-N ' -(4-(2-methylpropyloxy)pheny lmethy 1) carbamide in the range of about 6.0 ng/mL to about 40.0 ng/mL.
23. The use of Claim 20, wherein the amount of N-(l-methylpiperidin-4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the medicament results in a steady state blood plasma concentration of N-(l-methylpiperidin- 4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the range of about 8.5 ng/mL to about 35.0 ng/mL.
24. The use of Claim 20, wherein the medicament is suitable for administration once every day.
25. The use of Claim 20, wherein the amount of N-(l-methylpiperidin-4-yl)-N- (4-fluorophenylmethyl)-N>-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the medicament is in the range of about 0.5 mg to about 50 mg.
26. The use of Claim 20, wherein the amount of N-(l-methylpiperidin-4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the medicament is in the range of about 1 mg to about 40 mg.
27. The use of Claim 20, wherein the amount of N-( 1 -methylpiperidin-4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the medicament is in the range of about 2.5 mg to about 30 mg.
28. The use of Claim 20, wherein the amount of N-(l-methylpiperidin-4-yl)-N- (4-fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide in the medicament is in the range of about 5 mg to about 20 mg.
29. The use of Claim 20, wherein the N-(l-methylpiperidin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylm ethyl) carbamide does not affect a sleep parameter selected from the group consisting of sleep period time, total sleep time, sleep onset latency, number of stage shifts, total time awake, early morning wake, sleep efficiency index, microarousal index, and a REM sleep parameter.
30. The use of Claim 29, wherein the REM sleep parameter is selected from the group consisting of REM sleep duration, proportion of REM sleep, REM sleep latency, REM activity and REM density.
31. The use of Claim 20, wherein the medicament is provided in a first dosage form and a second dosage form of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)- N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide said first dosage form being adapted for administration at least 24 hours prior to administration of the second dosage form.
32. The use of Claim 31, wherein the first dosage form contains a higher dose of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide than the second dosage form.
33. The use of Claim 20, wherein the medicament is provided along with information indicating that N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4- (2-methylpropyloxy)phenylmethyl) carbamide increases slow wave sleep.
34. The use of Claim 33, wherein the information comprises printed matter that advises that N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide ameliorates insomnia.
35. The use of Claim 34, wherein said printed matter is a label.
36. The use of Claim 20, wherein the medicament is suitable for administration at a time other than immediately before a sleep period.
37. The use of Claim 36, wherein the medicament is suitable for administration in the morning.
38. Use of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide for the preparation of a medicament for decreasing the number of awakenings after sleep onset.
39. Use of N-(l-methylρiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide for the preparation of a medicament for decreasing the time awake after sleep onset.
40. A method of manufacturing a pharmaceutical composition, said method comprising: obtaining a first dosage form comprising a first amount of N-(I- methylpiperidin-4-yl)-N-(4-fiuorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide; obtaining a second dosage form comprising a second amount of N-(I- methylpiperidin~4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide; and packaging together said first dosage form and said second dosage form.
41. The method of Claim 40, wherein the first dosage form and the second dosage form each comprise an oral dosage form.
42. The method of Claim 40, wherein the first dosage form contains a higher dose of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide than the second dosage form.
43. A packaged pharmaceutical composition, comprising N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide in a container and instructions for using N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide to treat insomnia.
44. A packaged pharmaceutical composition, comprising N-(I- methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylproρyloxy)ρhenylmethyl) carbamide in a container and instructions for using N-(I- methylpiperidin-4-yi)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylρropyloxy)phenylmethyl) carbamide to increase slow wave sleep, decrease the number of awakenings after sleep onset or decrease the time awake after sleep onset.
45. A kit comprising a first dosage form of N-(l-methylρiρeridin-4-yl)-N-(4- fluorophenylmethyl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide and a second dosage form of N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylrnethyl) carbamide.
46. The kit of Claim 45, wherein the first dosage form contains a higher does of N-(I -methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide than the second dosage form.
47. The kit of Claim 45, wherein the kit further comprises instructions for taking the first dosage form at least 24 hours before taking the second dosage form.
48. The kit of Claim 45, wherein the instructions are on a label.
49. The kit of Claim 48, wherein the instructions further inform a subject that N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide increases slow wave sleep.
50. The kit of Claim 49, wherein the instructions further inform a subject that N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide ameliorates insomnia.
51. The kit of Claim 49, wherein the instructions further inform a subject that N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide decreases the number of awakenings after sleep onset.
52. The kit of Claim 49, wherein the instructions further inform a subject that N-(l-methylpiperidin-4-yl)-N-(4-fluorophenylmethyl)-N'-(4-(2- methylpropyloxy)phenylmethyl) carbamide decreases the time awake after sleep onset.
PCT/US2007/009804 2006-04-19 2007-04-18 Use of 4-amino-piperidines for treating sleep disorders WO2007124136A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US79374106P 2006-04-19 2006-04-19
US60/793,741 2006-04-19

Publications (1)

Publication Number Publication Date
WO2007124136A1 true WO2007124136A1 (en) 2007-11-01

Family

ID=38328553

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/009804 WO2007124136A1 (en) 2006-04-19 2007-04-18 Use of 4-amino-piperidines for treating sleep disorders

Country Status (1)

Country Link
WO (1) WO2007124136A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2699546A1 (en) * 2011-04-18 2014-02-26 Commonwealth Scientific & Industrial Research Organisation Gas capture process
CN105481757A (en) * 2015-12-25 2016-04-13 北京康立生医药技术开发有限公司 Preparation method of pimavanserin
CN105859608A (en) * 2016-05-27 2016-08-17 成都百裕制药股份有限公司 Method for preparing pimavanserin hemitartrate crystal form B
CN105906551A (en) * 2016-04-27 2016-08-31 东南大学 Preparation method of 5-HT2A inverse agonist ACP-103
CN106008323A (en) * 2016-05-30 2016-10-12 成都百裕制药股份有限公司 Preparation method of pimavanserin hemitartrate crystal form C
JP2017516819A (en) * 2014-06-02 2017-06-22 ラボラトリオス・デル・デエレ・エステベ・エセ・ア Alkyl and aryl derivatives of 1-oxa-4,9-diazaspiroundecane compounds having various activities against pain
JP2018522012A (en) * 2015-07-20 2018-08-09 アカディア ファーマシューティカルズ インコーポレーテッド Prepare N- (4-fluorobenzyl) -N- (1-methylpiperidin-4-yl) -N ′-(4- (2-methylpropyloxy) phenylmethyl) carbamide and its tartrate salt and polymorph Form C how to
CN109111385A (en) * 2017-06-23 2019-01-01 上海翰森生物医药科技有限公司 5-HT2AAcceptor inhibitor and its preparation method and application
IT201800003736A1 (en) * 2018-03-19 2019-09-19 Lundbeck Pharmaceuticals Italy S P A Process for the production of Pimavanserin
US10449185B2 (en) 2017-08-30 2019-10-22 Acadia Pharmaceuticals Inc. Formulations of pimavanserin
US10517860B2 (en) 2016-03-25 2019-12-31 Acadia Pharmaceuticals Inc. Combination of pimavanserin and cytochrome P450 modulators
WO2020048831A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 5-aryl-3,9-diazaspiro[5.5]undecan-2-one compounds
WO2020048826A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 5-substituted 1-oxa-3,9-diazaspiro[5.5]undecan-2-one compounds
WO2020048829A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 3,9-diazaspiro[5.5]undecane compounds
WO2020048827A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 1, 3, 9-triazaspiro[5.5] undecan-2-one compounds
WO2020048828A1 (en) 2018-09-03 2020-03-12 Bayer Pharma Aktiengesellschaft 5-heteroaryl-3,9-diazaspiro[5.5]undecane compounds
WO2020048830A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 5-aryl-3,9-diazaspiro[5.5]undecan-2-one compounds
US10689398B2 (en) 2015-10-23 2020-06-23 Esteve Pharmaceuticals, S.A. OXA-Diazaspiro compounds having activity against pain
US10953000B2 (en) 2016-03-25 2021-03-23 Acadia Pharmaceuticals Inc. Combination of pimavanserin and cytochrome P450 modulators
WO2021193790A1 (en) * 2020-03-26 2021-09-30 塩野義製薬株式会社 Aromatic heterocyclic derivative having serotonin receptor binding activity
US11135211B2 (en) 2017-04-28 2021-10-05 Acadia Pharmaceuticals Inc. Pimavanserin for treating impulse control disorder
US11464768B2 (en) 2016-12-20 2022-10-11 Acadia Pharmaceuticals Inc. Pimavanserin alone or in combination for use in the treatment of Alzheimer's disease psychosis

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6815458B2 (en) * 2000-03-06 2004-11-09 Acadia Pharmaceuticals, Inc Azacyclic compounds
WO2005112927A1 (en) * 2004-05-21 2005-12-01 Acadia Pharmaceuticals Inc. Selective serotonin receptor inverse agonists as therapeutics for disease
WO2006036874A1 (en) * 2004-09-27 2006-04-06 Acadia Pharmaceuticals Inc. Salts of n-(4-fluorobenzyl)-n-(1-methylpiperidin-4-yl)-n'-(4-(2-methylpropyloxy)phenylmethyl)carbamide and their preparation

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6815458B2 (en) * 2000-03-06 2004-11-09 Acadia Pharmaceuticals, Inc Azacyclic compounds
WO2005112927A1 (en) * 2004-05-21 2005-12-01 Acadia Pharmaceuticals Inc. Selective serotonin receptor inverse agonists as therapeutics for disease
WO2006036874A1 (en) * 2004-09-27 2006-04-06 Acadia Pharmaceuticals Inc. Salts of n-(4-fluorobenzyl)-n-(1-methylpiperidin-4-yl)-n'-(4-(2-methylpropyloxy)phenylmethyl)carbamide and their preparation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
"ACP-103", DRUGS OF THE FUTURE, vol. 31, no. 11, 2006 - 2006, pages 939 - 943, XP002446571 *

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2699546A1 (en) * 2011-04-18 2014-02-26 Commonwealth Scientific & Industrial Research Organisation Gas capture process
EP2699546A4 (en) * 2011-04-18 2014-09-24 Commw Scient Ind Res Org Gas capture process
US9409122B2 (en) 2011-04-18 2016-08-09 Commonwealth Scientific And Industrial Research Organisation Gas capture process
JP2017516819A (en) * 2014-06-02 2017-06-22 ラボラトリオス・デル・デエレ・エステベ・エセ・ア Alkyl and aryl derivatives of 1-oxa-4,9-diazaspiroundecane compounds having various activities against pain
US10703765B2 (en) * 2014-06-02 2020-07-07 Esteve Pharmaceuticals, S.A. Alkyl and aryl derivatives of 1-oxa-4,9-diazaspiro undecane compounds having multimodal activity against pain
JP2018522012A (en) * 2015-07-20 2018-08-09 アカディア ファーマシューティカルズ インコーポレーテッド Prepare N- (4-fluorobenzyl) -N- (1-methylpiperidin-4-yl) -N ′-(4- (2-methylpropyloxy) phenylmethyl) carbamide and its tartrate salt and polymorph Form C how to
US10981870B2 (en) 2015-07-20 2021-04-20 Acadia Pharmaceuticals Inc. Methods for preparing N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and its tartrate salt and polymorphic form
US11840515B2 (en) 2015-07-20 2023-12-12 Acadia Pharmaceuticals Inc. Methods for preparing N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and its tartrate salt and polymorphic form c
US10981871B2 (en) 2015-07-20 2021-04-20 Acadia Pharmaceuticals Inc. Methods for preparing N-(4-fluorobenzyl)-N-(1-methylpiperidin-4-yl)-N′-(4-(2-methylpropyloxy)phenylmethyl)carbamide and its tartrate salt and polymorphic form C
US10689398B2 (en) 2015-10-23 2020-06-23 Esteve Pharmaceuticals, S.A. OXA-Diazaspiro compounds having activity against pain
CN105481757A (en) * 2015-12-25 2016-04-13 北京康立生医药技术开发有限公司 Preparation method of pimavanserin
US10517860B2 (en) 2016-03-25 2019-12-31 Acadia Pharmaceuticals Inc. Combination of pimavanserin and cytochrome P450 modulators
US10953000B2 (en) 2016-03-25 2021-03-23 Acadia Pharmaceuticals Inc. Combination of pimavanserin and cytochrome P450 modulators
US11191757B2 (en) 2016-03-25 2021-12-07 Acadia Pharmaceuticals Inc. Combination of pimavanserin and cytochrome P450 modulators
CN105906551A (en) * 2016-04-27 2016-08-31 东南大学 Preparation method of 5-HT2A inverse agonist ACP-103
CN105859608A (en) * 2016-05-27 2016-08-17 成都百裕制药股份有限公司 Method for preparing pimavanserin hemitartrate crystal form B
CN106008323B (en) * 2016-05-30 2018-11-23 成都百裕制药股份有限公司 A method of preparing half tartrate crystal form C of piperazine Ma Selin
CN106008323A (en) * 2016-05-30 2016-10-12 成都百裕制药股份有限公司 Preparation method of pimavanserin hemitartrate crystal form C
US11464768B2 (en) 2016-12-20 2022-10-11 Acadia Pharmaceuticals Inc. Pimavanserin alone or in combination for use in the treatment of Alzheimer's disease psychosis
US11135211B2 (en) 2017-04-28 2021-10-05 Acadia Pharmaceuticals Inc. Pimavanserin for treating impulse control disorder
CN109111385B (en) * 2017-06-23 2023-06-30 上海翰森生物医药科技有限公司 5-HT 2A Receptor inhibitor and preparation method and application thereof
CN109111385A (en) * 2017-06-23 2019-01-01 上海翰森生物医药科技有限公司 5-HT2AAcceptor inhibitor and its preparation method and application
US10849891B2 (en) 2017-08-30 2020-12-01 Acadia Pharmaceuticals Inc. Formulations of pimavanserin
US10646480B2 (en) 2017-08-30 2020-05-12 Acadia Pharmaceuticals Inc. Formulations of pimavanserin
US11452721B2 (en) 2017-08-30 2022-09-27 Acadia Pharmaceuticals Inc. Formulations of pimavanserin
US10449185B2 (en) 2017-08-30 2019-10-22 Acadia Pharmaceuticals Inc. Formulations of pimavanserin
WO2019179920A1 (en) * 2018-03-19 2019-09-26 Lundbeck Pharmaceuticals Italy S.P.A. Process for the manufacturing of pimavanserin
IT201800003736A1 (en) * 2018-03-19 2019-09-19 Lundbeck Pharmaceuticals Italy S P A Process for the production of Pimavanserin
WO2020048830A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 5-aryl-3,9-diazaspiro[5.5]undecan-2-one compounds
WO2020048828A1 (en) 2018-09-03 2020-03-12 Bayer Pharma Aktiengesellschaft 5-heteroaryl-3,9-diazaspiro[5.5]undecane compounds
WO2020048827A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 1, 3, 9-triazaspiro[5.5] undecan-2-one compounds
WO2020048829A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 3,9-diazaspiro[5.5]undecane compounds
WO2020048826A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 5-substituted 1-oxa-3,9-diazaspiro[5.5]undecan-2-one compounds
WO2020048831A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 5-aryl-3,9-diazaspiro[5.5]undecan-2-one compounds
WO2021193790A1 (en) * 2020-03-26 2021-09-30 塩野義製薬株式会社 Aromatic heterocyclic derivative having serotonin receptor binding activity

Similar Documents

Publication Publication Date Title
WO2007124136A1 (en) Use of 4-amino-piperidines for treating sleep disorders
RU2320646C2 (en) N-substituted derivatives of piperidine as serotonin receptor agents
US7538222B2 (en) N-substituted piperidine derivatives as serotonin receptor agents
US7351707B2 (en) Spiroazacyclic compounds as monoamine receptor modulators
US7253186B2 (en) N-substituted piperidine derivatives as serotonin receptor agents
US20080051429A1 (en) Use of 4-amino-piperidines for treating sleep disorders
US20030220316A1 (en) Azacyclic compounds
EA021755B1 (en) PYRAZINE DERIVATIVES AS ENaC BLOCKERS
KR20230002483A (en) BCL-2 protein inhibitor
EP1444200B1 (en) Azetidinyl diamines useful as ligands of the nociceptin receptor orl-1
AU2007203444B2 (en) N-substituted piperidine derivatives as serotonin receptor agents
AU2002361872C1 (en) Spiroazacyclic compounds as monoamine receptor modulators
EP1787984B1 (en) Azacyclic compounds for use in the treatment of serotonin related diseases

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07755888

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07755888

Country of ref document: EP

Kind code of ref document: A1